WO2022097716A1 - Method for producing reversibly immortalized cell - Google Patents
Method for producing reversibly immortalized cell Download PDFInfo
- Publication number
- WO2022097716A1 WO2022097716A1 PCT/JP2021/040757 JP2021040757W WO2022097716A1 WO 2022097716 A1 WO2022097716 A1 WO 2022097716A1 JP 2021040757 W JP2021040757 W JP 2021040757W WO 2022097716 A1 WO2022097716 A1 WO 2022097716A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- cells
- gene
- cell
- vector
- immortalized
- Prior art date
Links
- 238000004519 manufacturing process Methods 0.000 title claims abstract description 16
- 210000004027 cell Anatomy 0.000 claims abstract description 476
- 239000013598 vector Substances 0.000 claims abstract description 188
- 108090000623 proteins and genes Proteins 0.000 claims abstract description 143
- 101100058550 Mus musculus Bmi1 gene Proteins 0.000 claims abstract description 65
- 238000000034 method Methods 0.000 claims abstract description 50
- 101150047500 TERT gene Proteins 0.000 claims abstract description 27
- 238000012258 culturing Methods 0.000 claims abstract description 24
- 241001493065 dsRNA viruses Species 0.000 claims abstract description 16
- 210000004962 mammalian cell Anatomy 0.000 claims abstract description 5
- 241000711408 Murine respirovirus Species 0.000 claims description 156
- 210000002901 mesenchymal stem cell Anatomy 0.000 claims description 133
- 239000003814 drug Substances 0.000 claims description 19
- 230000002759 chromosomal effect Effects 0.000 claims description 16
- 230000001172 regenerating effect Effects 0.000 claims description 15
- 239000013603 viral vector Substances 0.000 claims description 14
- 238000010367 cloning Methods 0.000 claims description 13
- 230000002441 reversible effect Effects 0.000 claims description 12
- 210000001082 somatic cell Anatomy 0.000 claims description 10
- 210000001988 somatic stem cell Anatomy 0.000 claims description 6
- 230000008569 process Effects 0.000 claims description 2
- 210000000349 chromosome Anatomy 0.000 abstract description 30
- 208000015181 infectious disease Diseases 0.000 description 75
- 239000002609 medium Substances 0.000 description 71
- 101000965899 Simian virus 40 Large T antigen Proteins 0.000 description 50
- 230000004069 differentiation Effects 0.000 description 47
- 241000700605 Viruses Species 0.000 description 28
- 238000004458 analytical method Methods 0.000 description 28
- 230000004663 cell proliferation Effects 0.000 description 28
- 230000014509 gene expression Effects 0.000 description 23
- 230000001939 inductive effect Effects 0.000 description 20
- 108091035539 telomere Proteins 0.000 description 20
- 210000003411 telomere Anatomy 0.000 description 20
- 102000055501 telomere Human genes 0.000 description 20
- 230000012010 growth Effects 0.000 description 18
- 239000000047 product Substances 0.000 description 18
- 210000002569 neuron Anatomy 0.000 description 17
- 108010017842 Telomerase Proteins 0.000 description 16
- 230000035755 proliferation Effects 0.000 description 16
- 238000012546 transfer Methods 0.000 description 15
- 210000001789 adipocyte Anatomy 0.000 description 14
- 230000000694 effects Effects 0.000 description 14
- 230000035772 mutation Effects 0.000 description 13
- 210000000130 stem cell Anatomy 0.000 description 13
- 102100032938 Telomerase reverse transcriptase Human genes 0.000 description 12
- 238000010370 cell cloning Methods 0.000 description 12
- 230000032823 cell division Effects 0.000 description 12
- 210000000963 osteoblast Anatomy 0.000 description 11
- 102100036011 T-cell surface glycoprotein CD4 Human genes 0.000 description 9
- 210000001612 chondrocyte Anatomy 0.000 description 9
- 210000004700 fetal blood Anatomy 0.000 description 9
- 210000002966 serum Anatomy 0.000 description 9
- 238000012360 testing method Methods 0.000 description 9
- 108020004414 DNA Proteins 0.000 description 8
- 241000700159 Rattus Species 0.000 description 8
- 238000004113 cell culture Methods 0.000 description 8
- 230000008859 change Effects 0.000 description 8
- 239000013641 positive control Substances 0.000 description 8
- 230000002062 proliferating effect Effects 0.000 description 8
- 210000001519 tissue Anatomy 0.000 description 8
- 238000002054 transplantation Methods 0.000 description 8
- 230000024245 cell differentiation Effects 0.000 description 7
- 230000007423 decrease Effects 0.000 description 7
- -1 etc.) Proteins 0.000 description 7
- 238000002474 experimental method Methods 0.000 description 7
- 239000013642 negative control Substances 0.000 description 7
- 102000004169 proteins and genes Human genes 0.000 description 7
- 239000012679 serum free medium Substances 0.000 description 7
- 238000010186 staining Methods 0.000 description 7
- 239000006144 Dulbecco’s modified Eagle's medium Substances 0.000 description 6
- 239000000853 adhesive Substances 0.000 description 6
- 230000001070 adhesive effect Effects 0.000 description 6
- 210000001185 bone marrow Anatomy 0.000 description 6
- 230000003247 decreasing effect Effects 0.000 description 6
- 230000006870 function Effects 0.000 description 6
- 230000007774 longterm Effects 0.000 description 6
- 239000000243 solution Substances 0.000 description 6
- UCSJYZPVAKXKNQ-HZYVHMACSA-N streptomycin Chemical compound CN[C@H]1[C@H](O)[C@@H](O)[C@H](CO)O[C@H]1O[C@@H]1[C@](C=O)(O)[C@H](C)O[C@H]1O[C@@H]1[C@@H](NC(N)=N)[C@H](O)[C@@H](NC(N)=N)[C@H](O)[C@H]1O UCSJYZPVAKXKNQ-HZYVHMACSA-N 0.000 description 6
- 101001065501 Escherichia phage MS2 Lysis protein Proteins 0.000 description 5
- 101000958041 Homo sapiens Musculin Proteins 0.000 description 5
- 241001465754 Metazoa Species 0.000 description 5
- 210000000577 adipose tissue Anatomy 0.000 description 5
- 230000032683 aging Effects 0.000 description 5
- 238000012790 confirmation Methods 0.000 description 5
- 239000007850 fluorescent dye Substances 0.000 description 5
- 102000046949 human MSC Human genes 0.000 description 5
- 230000006698 induction Effects 0.000 description 5
- 238000012423 maintenance Methods 0.000 description 5
- 239000000203 mixture Substances 0.000 description 5
- 239000002245 particle Substances 0.000 description 5
- 238000011144 upstream manufacturing Methods 0.000 description 5
- CIWBSHSKHKDKBQ-JLAZNSOCSA-N Ascorbic acid Chemical compound OC[C@H](O)[C@H]1OC(=O)C(O)=C1O CIWBSHSKHKDKBQ-JLAZNSOCSA-N 0.000 description 4
- 208000003322 Coinfection Diseases 0.000 description 4
- 102000004190 Enzymes Human genes 0.000 description 4
- 108090000790 Enzymes Proteins 0.000 description 4
- 101150034814 F gene Proteins 0.000 description 4
- 101150008820 HN gene Proteins 0.000 description 4
- 101001057504 Homo sapiens Interferon-stimulated gene 20 kDa protein Proteins 0.000 description 4
- 101001055144 Homo sapiens Interleukin-2 receptor subunit alpha Proteins 0.000 description 4
- 102100027268 Interferon-stimulated gene 20 kDa protein Human genes 0.000 description 4
- 101710181008 P protein Proteins 0.000 description 4
- 101710177166 Phosphoprotein Proteins 0.000 description 4
- 241000711931 Rhabdoviridae Species 0.000 description 4
- 108010081734 Ribonucleoproteins Proteins 0.000 description 4
- 102000004389 Ribonucleoproteins Human genes 0.000 description 4
- 210000004102 animal cell Anatomy 0.000 description 4
- 210000004748 cultured cell Anatomy 0.000 description 4
- 235000021186 dishes Nutrition 0.000 description 4
- 210000002950 fibroblast Anatomy 0.000 description 4
- 210000004263 induced pluripotent stem cell Anatomy 0.000 description 4
- NOESYZHRGYRDHS-UHFFFAOYSA-N insulin Chemical compound N1C(=O)C(NC(=O)C(CCC(N)=O)NC(=O)C(CCC(O)=O)NC(=O)C(C(C)C)NC(=O)C(NC(=O)CN)C(C)CC)CSSCC(C(NC(CO)C(=O)NC(CC(C)C)C(=O)NC(CC=2C=CC(O)=CC=2)C(=O)NC(CCC(N)=O)C(=O)NC(CC(C)C)C(=O)NC(CCC(O)=O)C(=O)NC(CC(N)=O)C(=O)NC(CC=2C=CC(O)=CC=2)C(=O)NC(CSSCC(NC(=O)C(C(C)C)NC(=O)C(CC(C)C)NC(=O)C(CC=2C=CC(O)=CC=2)NC(=O)C(CC(C)C)NC(=O)C(C)NC(=O)C(CCC(O)=O)NC(=O)C(C(C)C)NC(=O)C(CC(C)C)NC(=O)C(CC=2NC=NC=2)NC(=O)C(CO)NC(=O)CNC2=O)C(=O)NCC(=O)NC(CCC(O)=O)C(=O)NC(CCCNC(N)=N)C(=O)NCC(=O)NC(CC=3C=CC=CC=3)C(=O)NC(CC=3C=CC=CC=3)C(=O)NC(CC=3C=CC(O)=CC=3)C(=O)NC(C(C)O)C(=O)N3C(CCC3)C(=O)NC(CCCCN)C(=O)NC(C)C(O)=O)C(=O)NC(CC(N)=O)C(O)=O)=O)NC(=O)C(C(C)CC)NC(=O)C(CO)NC(=O)C(C(C)O)NC(=O)C1CSSCC2NC(=O)C(CC(C)C)NC(=O)C(NC(=O)C(CCC(N)=O)NC(=O)C(CC(N)=O)NC(=O)C(NC(=O)C(N)CC=1C=CC=CC=1)C(C)C)CC1=CN=CN1 NOESYZHRGYRDHS-UHFFFAOYSA-N 0.000 description 4
- 239000011159 matrix material Substances 0.000 description 4
- 210000005036 nerve Anatomy 0.000 description 4
- 102000039446 nucleic acids Human genes 0.000 description 4
- 108020004707 nucleic acids Proteins 0.000 description 4
- 150000007523 nucleic acids Chemical class 0.000 description 4
- 230000004072 osteoblast differentiation Effects 0.000 description 4
- 230000002035 prolonged effect Effects 0.000 description 4
- 238000003908 quality control method Methods 0.000 description 4
- 230000002829 reductive effect Effects 0.000 description 4
- 108010057210 telomerase RNA Proteins 0.000 description 4
- 102000002260 Alkaline Phosphatase Human genes 0.000 description 3
- 108020004774 Alkaline Phosphatase Proteins 0.000 description 3
- 102000008186 Collagen Human genes 0.000 description 3
- 108010035532 Collagen Proteins 0.000 description 3
- 208000004232 Enteritis Diseases 0.000 description 3
- 102000018233 Fibroblast Growth Factor Human genes 0.000 description 3
- 108050007372 Fibroblast Growth Factor Proteins 0.000 description 3
- 108090000379 Fibroblast growth factor 2 Proteins 0.000 description 3
- 102100024785 Fibroblast growth factor 2 Human genes 0.000 description 3
- 102000034615 Glial cell line-derived neurotrophic factor Human genes 0.000 description 3
- 108091010837 Glial cell line-derived neurotrophic factor Proteins 0.000 description 3
- PEDCQBHIVMGVHV-UHFFFAOYSA-N Glycerine Chemical compound OCC(O)CO PEDCQBHIVMGVHV-UHFFFAOYSA-N 0.000 description 3
- 108090000723 Insulin-Like Growth Factor I Proteins 0.000 description 3
- 102000004218 Insulin-Like Growth Factor I Human genes 0.000 description 3
- 101150118742 NP gene Proteins 0.000 description 3
- 206010028980 Neoplasm Diseases 0.000 description 3
- 102100034574 P protein Human genes 0.000 description 3
- 241000711504 Paramyxoviridae Species 0.000 description 3
- 229930182555 Penicillin Natural products 0.000 description 3
- JGSARLDLIJGVTE-MBNYWOFBSA-N Penicillin G Chemical compound N([C@H]1[C@H]2SC([C@@H](N2C1=O)C(O)=O)(C)C)C(=O)CC1=CC=CC=C1 JGSARLDLIJGVTE-MBNYWOFBSA-N 0.000 description 3
- 108010000598 Polycomb Repressive Complex 1 Proteins 0.000 description 3
- 102000002273 Polycomb Repressive Complex 1 Human genes 0.000 description 3
- 230000009815 adipogenic differentiation Effects 0.000 description 3
- 230000037396 body weight Effects 0.000 description 3
- 210000003321 cartilage cell Anatomy 0.000 description 3
- 229920001436 collagen Polymers 0.000 description 3
- 210000000805 cytoplasm Anatomy 0.000 description 3
- 210000002889 endothelial cell Anatomy 0.000 description 3
- 229940126864 fibroblast growth factor Drugs 0.000 description 3
- 230000004927 fusion Effects 0.000 description 3
- 101150082979 gdnf gene Proteins 0.000 description 3
- 210000000688 human artificial chromosome Anatomy 0.000 description 3
- 230000002458 infectious effect Effects 0.000 description 3
- 238000003780 insertion Methods 0.000 description 3
- 230000037431 insertion Effects 0.000 description 3
- 210000001165 lymph node Anatomy 0.000 description 3
- 239000012528 membrane Substances 0.000 description 3
- 230000009818 osteogenic differentiation Effects 0.000 description 3
- 229940049954 penicillin Drugs 0.000 description 3
- 238000002360 preparation method Methods 0.000 description 3
- 230000006798 recombination Effects 0.000 description 3
- 230000001105 regulatory effect Effects 0.000 description 3
- 238000011160 research Methods 0.000 description 3
- 210000000952 spleen Anatomy 0.000 description 3
- 229960005322 streptomycin Drugs 0.000 description 3
- 210000003954 umbilical cord Anatomy 0.000 description 3
- YBJHBAHKTGYVGT-ZKWXMUAHSA-N (+)-Biotin Chemical compound N1C(=O)N[C@@H]2[C@H](CCCCC(=O)O)SC[C@@H]21 YBJHBAHKTGYVGT-ZKWXMUAHSA-N 0.000 description 2
- JKMHFZQWWAIEOD-UHFFFAOYSA-N 2-[4-(2-hydroxyethyl)piperazin-1-yl]ethanesulfonic acid Chemical compound OCC[NH+]1CCN(CCS([O-])(=O)=O)CC1 JKMHFZQWWAIEOD-UHFFFAOYSA-N 0.000 description 2
- FWMNVWWHGCHHJJ-SKKKGAJSSA-N 4-amino-1-[(2r)-6-amino-2-[[(2r)-2-[[(2r)-2-[[(2r)-2-amino-3-phenylpropanoyl]amino]-3-phenylpropanoyl]amino]-4-methylpentanoyl]amino]hexanoyl]piperidine-4-carboxylic acid Chemical compound C([C@H](C(=O)N[C@H](CC(C)C)C(=O)N[C@H](CCCCN)C(=O)N1CCC(N)(CC1)C(O)=O)NC(=O)[C@H](N)CC=1C=CC=CC=1)C1=CC=CC=C1 FWMNVWWHGCHHJJ-SKKKGAJSSA-N 0.000 description 2
- 241000283690 Bos taurus Species 0.000 description 2
- 102000004219 Brain-derived neurotrophic factor Human genes 0.000 description 2
- 108090000715 Brain-derived neurotrophic factor Proteins 0.000 description 2
- 208000031404 Chromosome Aberrations Diseases 0.000 description 2
- 235000005956 Cosmos caudatus Nutrition 0.000 description 2
- 208000000655 Distemper Diseases 0.000 description 2
- 241000282326 Felis catus Species 0.000 description 2
- 102000016359 Fibronectins Human genes 0.000 description 2
- 108010067306 Fibronectins Proteins 0.000 description 2
- WSFSSNUMVMOOMR-UHFFFAOYSA-N Formaldehyde Chemical compound O=C WSFSSNUMVMOOMR-UHFFFAOYSA-N 0.000 description 2
- DHCLVCXQIBBOPH-UHFFFAOYSA-N Glycerol 2-phosphate Chemical compound OCC(CO)OP(O)(O)=O DHCLVCXQIBBOPH-UHFFFAOYSA-N 0.000 description 2
- 241000282412 Homo Species 0.000 description 2
- 101000655352 Homo sapiens Telomerase reverse transcriptase Proteins 0.000 description 2
- 101150088952 IGF1 gene Proteins 0.000 description 2
- 208000022559 Inflammatory bowel disease Diseases 0.000 description 2
- 102000004877 Insulin Human genes 0.000 description 2
- 108090001061 Insulin Proteins 0.000 description 2
- 239000007760 Iscove's Modified Dulbecco's Medium Substances 0.000 description 2
- 108060001084 Luciferase Proteins 0.000 description 2
- 239000005089 Luciferase Substances 0.000 description 2
- 206010025323 Lymphomas Diseases 0.000 description 2
- 241000712079 Measles morbillivirus Species 0.000 description 2
- 241000699666 Mus <mouse, genus> Species 0.000 description 2
- 241000209094 Oryza Species 0.000 description 2
- 235000007164 Oryza sativa Nutrition 0.000 description 2
- 238000010222 PCR analysis Methods 0.000 description 2
- RJKFOVLPORLFTN-LEKSSAKUSA-N Progesterone Chemical compound C1CC2=CC(=O)CC[C@]2(C)[C@@H]2[C@@H]1[C@@H]1CC[C@H](C(=O)C)[C@@]1(C)CC2 RJKFOVLPORLFTN-LEKSSAKUSA-N 0.000 description 2
- 108091081021 Sense strand Proteins 0.000 description 2
- 210000001744 T-lymphocyte Anatomy 0.000 description 2
- 102100033254 Tumor suppressor ARF Human genes 0.000 description 2
- 241000711975 Vesicular stomatitis virus Species 0.000 description 2
- 108010067390 Viral Proteins Proteins 0.000 description 2
- 230000003698 anagen phase Effects 0.000 description 2
- 230000000692 anti-sense effect Effects 0.000 description 2
- 229960005070 ascorbic acid Drugs 0.000 description 2
- 235000010323 ascorbic acid Nutrition 0.000 description 2
- 239000011668 ascorbic acid Substances 0.000 description 2
- 239000007640 basal medium Substances 0.000 description 2
- 230000015572 biosynthetic process Effects 0.000 description 2
- 210000004369 blood Anatomy 0.000 description 2
- 239000008280 blood Substances 0.000 description 2
- 229940077737 brain-derived neurotrophic factor Drugs 0.000 description 2
- 238000004364 calculation method Methods 0.000 description 2
- 210000000845 cartilage Anatomy 0.000 description 2
- 230000032677 cell aging Effects 0.000 description 2
- 239000006143 cell culture medium Substances 0.000 description 2
- 230000007910 cell fusion Effects 0.000 description 2
- 230000010094 cellular senescence Effects 0.000 description 2
- 238000006243 chemical reaction Methods 0.000 description 2
- 230000001276 controlling effect Effects 0.000 description 2
- 238000012136 culture method Methods 0.000 description 2
- 238000001514 detection method Methods 0.000 description 2
- 201000010099 disease Diseases 0.000 description 2
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 2
- 239000006185 dispersion Substances 0.000 description 2
- 235000020776 essential amino acid Nutrition 0.000 description 2
- 239000003797 essential amino acid Substances 0.000 description 2
- 238000011156 evaluation Methods 0.000 description 2
- 108091006047 fluorescent proteins Proteins 0.000 description 2
- 102000034287 fluorescent proteins Human genes 0.000 description 2
- 238000005194 fractionation Methods 0.000 description 2
- 238000007710 freezing Methods 0.000 description 2
- 230000008014 freezing Effects 0.000 description 2
- 210000001035 gastrointestinal tract Anatomy 0.000 description 2
- 230000035876 healing Effects 0.000 description 2
- 210000003958 hematopoietic stem cell Anatomy 0.000 description 2
- 210000003494 hepatocyte Anatomy 0.000 description 2
- JYGXADMDTFJGBT-VWUMJDOOSA-N hydrocortisone Chemical compound O=C1CC[C@]2(C)[C@H]3[C@@H](O)C[C@](C)([C@@](CC4)(O)C(=O)CO)[C@@H]4[C@@H]3CCC2=C1 JYGXADMDTFJGBT-VWUMJDOOSA-N 0.000 description 2
- CGIGDMFJXJATDK-UHFFFAOYSA-N indomethacin Chemical compound CC1=C(CC(O)=O)C2=CC(OC)=CC=C2N1C(=O)C1=CC=C(Cl)C=C1 CGIGDMFJXJATDK-UHFFFAOYSA-N 0.000 description 2
- 238000002347 injection Methods 0.000 description 2
- 239000007924 injection Substances 0.000 description 2
- 229940125396 insulin Drugs 0.000 description 2
- 238000002955 isolation Methods 0.000 description 2
- 150000002632 lipids Chemical class 0.000 description 2
- 230000007246 mechanism Effects 0.000 description 2
- 230000002107 myocardial effect Effects 0.000 description 2
- 230000001537 neural effect Effects 0.000 description 2
- 230000001272 neurogenic effect Effects 0.000 description 2
- 238000010899 nucleation Methods 0.000 description 2
- 239000013612 plasmid Substances 0.000 description 2
- 238000011002 quantification Methods 0.000 description 2
- 238000003753 real-time PCR Methods 0.000 description 2
- 238000005215 recombination Methods 0.000 description 2
- 230000010076 replication Effects 0.000 description 2
- 230000001177 retroviral effect Effects 0.000 description 2
- 235000009566 rice Nutrition 0.000 description 2
- 239000000523 sample Substances 0.000 description 2
- 230000009758 senescence Effects 0.000 description 2
- 238000004904 shortening Methods 0.000 description 2
- 210000004003 subcutaneous fat Anatomy 0.000 description 2
- 239000000758 substrate Substances 0.000 description 2
- 238000004114 suspension culture Methods 0.000 description 2
- 230000001225 therapeutic effect Effects 0.000 description 2
- 210000003606 umbilical vein Anatomy 0.000 description 2
- 241000712461 unidentified influenza virus Species 0.000 description 2
- 210000003462 vein Anatomy 0.000 description 2
- VUDQSRFCCHQIIU-UHFFFAOYSA-N 1-(3,5-dichloro-2,6-dihydroxy-4-methoxyphenyl)hexan-1-one Chemical compound CCCCCC(=O)C1=C(O)C(Cl)=C(OC)C(Cl)=C1O VUDQSRFCCHQIIU-UHFFFAOYSA-N 0.000 description 1
- NHBKXEKEPDILRR-UHFFFAOYSA-N 2,3-bis(butanoylsulfanyl)propyl butanoate Chemical compound CCCC(=O)OCC(SC(=O)CCC)CSC(=O)CCC NHBKXEKEPDILRR-UHFFFAOYSA-N 0.000 description 1
- APIXJSLKIYYUKG-UHFFFAOYSA-N 3 Isobutyl 1 methylxanthine Chemical compound O=C1N(C)C(=O)N(CC(C)C)C2=C1N=CN2 APIXJSLKIYYUKG-UHFFFAOYSA-N 0.000 description 1
- FWBHETKCLVMNFS-UHFFFAOYSA-N 4',6-Diamino-2-phenylindol Chemical compound C1=CC(C(=N)N)=CC=C1C1=CC2=CC=C(C(N)=N)C=C2N1 FWBHETKCLVMNFS-UHFFFAOYSA-N 0.000 description 1
- 102100027211 Albumin Human genes 0.000 description 1
- 108010088751 Albumins Proteins 0.000 description 1
- 206010002091 Anaesthesia Diseases 0.000 description 1
- 241000711404 Avian avulavirus 1 Species 0.000 description 1
- 239000012583 B-27 Supplement Substances 0.000 description 1
- 208000031648 Body Weight Changes Diseases 0.000 description 1
- 102100024506 Bone morphogenetic protein 2 Human genes 0.000 description 1
- 102100024505 Bone morphogenetic protein 4 Human genes 0.000 description 1
- 101000741929 Caenorhabditis elegans Serine/threonine-protein phosphatase 2A catalytic subunit Proteins 0.000 description 1
- 241000282472 Canis lupus familiaris Species 0.000 description 1
- 208000005623 Carcinogenesis Diseases 0.000 description 1
- 241000700198 Cavia Species 0.000 description 1
- 108010077544 Chromatin Proteins 0.000 description 1
- KRKNYBCHXYNGOX-UHFFFAOYSA-K Citrate Chemical compound [O-]C(=O)CC(O)(CC([O-])=O)C([O-])=O KRKNYBCHXYNGOX-UHFFFAOYSA-K 0.000 description 1
- 241000699800 Cricetinae Species 0.000 description 1
- KDXKERNSBIXSRK-RXMQYKEDSA-N D-lysine Chemical compound NCCCC[C@@H](N)C(O)=O KDXKERNSBIXSRK-RXMQYKEDSA-N 0.000 description 1
- 238000007400 DNA extraction Methods 0.000 description 1
- 208000010772 Dog disease Diseases 0.000 description 1
- 101150071673 E6 gene Proteins 0.000 description 1
- 101150013359 E7 gene Proteins 0.000 description 1
- 241001455610 Ephemerovirus Species 0.000 description 1
- 241000283086 Equidae Species 0.000 description 1
- 108010037362 Extracellular Matrix Proteins Proteins 0.000 description 1
- 102000010834 Extracellular Matrix Proteins Human genes 0.000 description 1
- 241000711950 Filoviridae Species 0.000 description 1
- 108010010803 Gelatin Proteins 0.000 description 1
- 229920002971 Heparan sulfate Polymers 0.000 description 1
- HTTJABKRGRZYRN-UHFFFAOYSA-N Heparin Chemical compound OC1C(NC(=O)C)C(O)OC(COS(O)(=O)=O)C1OC1C(OS(O)(=O)=O)C(O)C(OC2C(C(OS(O)(=O)=O)C(OC3C(C(O)C(O)C(O3)C(O)=O)OS(O)(=O)=O)C(CO)O2)NS(O)(=O)=O)C(C(O)=O)O1 HTTJABKRGRZYRN-UHFFFAOYSA-N 0.000 description 1
- 108010033040 Histones Proteins 0.000 description 1
- 108700005087 Homeobox Genes Proteins 0.000 description 1
- 101000762366 Homo sapiens Bone morphogenetic protein 2 Proteins 0.000 description 1
- 101000762379 Homo sapiens Bone morphogenetic protein 4 Proteins 0.000 description 1
- 101000980932 Homo sapiens Cyclin-dependent kinase inhibitor 2A Proteins 0.000 description 1
- 101001052035 Homo sapiens Fibroblast growth factor 2 Proteins 0.000 description 1
- 101000942967 Homo sapiens Leukemia inhibitory factor Proteins 0.000 description 1
- 101100313319 Homo sapiens TERT gene Proteins 0.000 description 1
- 101000954493 Human papillomavirus type 16 Protein E6 Proteins 0.000 description 1
- 102000017182 Ikaros Transcription Factor Human genes 0.000 description 1
- 108010013958 Ikaros Transcription Factor Proteins 0.000 description 1
- 102000015696 Interleukins Human genes 0.000 description 1
- 108010063738 Interleukins Proteins 0.000 description 1
- 101150062031 L gene Proteins 0.000 description 1
- 101710128836 Large T antigen Proteins 0.000 description 1
- 102100032352 Leukemia inhibitory factor Human genes 0.000 description 1
- 241000711828 Lyssavirus Species 0.000 description 1
- 101150039798 MYC gene Proteins 0.000 description 1
- 241000124008 Mammalia Species 0.000 description 1
- 241000712045 Morbillivirus Species 0.000 description 1
- 241000711386 Mumps virus Species 0.000 description 1
- 101100450263 Mus musculus Hadh gene Proteins 0.000 description 1
- 241000699670 Mus sp. Species 0.000 description 1
- 239000012580 N-2 Supplement Substances 0.000 description 1
- 208000009869 Neu-Laxova syndrome Diseases 0.000 description 1
- 102000005348 Neuraminidase Human genes 0.000 description 1
- 108010006232 Neuraminidase Proteins 0.000 description 1
- 108010077850 Nuclear Localization Signals Proteins 0.000 description 1
- 108091028043 Nucleic acid sequence Proteins 0.000 description 1
- 108700026244 Open Reading Frames Proteins 0.000 description 1
- 241000712464 Orthomyxoviridae Species 0.000 description 1
- 241000283973 Oryctolagus cuniculus Species 0.000 description 1
- 101150084044 P gene Proteins 0.000 description 1
- 239000012807 PCR reagent Substances 0.000 description 1
- 208000002606 Paramyxoviridae Infections Diseases 0.000 description 1
- 241000711902 Pneumovirus Species 0.000 description 1
- 101150040459 RAS gene Proteins 0.000 description 1
- 239000012980 RPMI-1640 medium Substances 0.000 description 1
- 238000011529 RT qPCR Methods 0.000 description 1
- 241000711798 Rabies lyssavirus Species 0.000 description 1
- 108091081062 Repeated sequence (DNA) Proteins 0.000 description 1
- 241000725643 Respiratory syncytial virus Species 0.000 description 1
- 241001113283 Respirovirus Species 0.000 description 1
- 241000711897 Rinderpest morbillivirus Species 0.000 description 1
- 241001533467 Rubulavirus Species 0.000 description 1
- 238000011579 SCID mouse model Methods 0.000 description 1
- 101710185500 Small t antigen Proteins 0.000 description 1
- 241000282887 Suidae Species 0.000 description 1
- 102000004357 Transferases Human genes 0.000 description 1
- 108090000992 Transferases Proteins 0.000 description 1
- 102000056172 Transforming growth factor beta-3 Human genes 0.000 description 1
- 108090000097 Transforming growth factor beta-3 Proteins 0.000 description 1
- 108700019146 Transgenes Proteins 0.000 description 1
- 102000044209 Tumor Suppressor Genes Human genes 0.000 description 1
- 108700025716 Tumor Suppressor Genes Proteins 0.000 description 1
- 108700005077 Viral Genes Proteins 0.000 description 1
- 235000010724 Wisteria floribunda Nutrition 0.000 description 1
- 230000005856 abnormality Effects 0.000 description 1
- 230000009471 action Effects 0.000 description 1
- 230000004913 activation Effects 0.000 description 1
- 230000001919 adrenal effect Effects 0.000 description 1
- 230000002411 adverse Effects 0.000 description 1
- 230000000735 allogeneic effect Effects 0.000 description 1
- 230000037005 anaesthesia Effects 0.000 description 1
- 239000003242 anti bacterial agent Substances 0.000 description 1
- 229940088710 antibiotic agent Drugs 0.000 description 1
- 108091007433 antigens Proteins 0.000 description 1
- 210000000436 anus Anatomy 0.000 description 1
- 238000013528 artificial neural network Methods 0.000 description 1
- 238000003149 assay kit Methods 0.000 description 1
- 230000002238 attenuated effect Effects 0.000 description 1
- 210000003719 b-lymphocyte Anatomy 0.000 description 1
- 230000033228 biological regulation Effects 0.000 description 1
- 230000005540 biological transmission Effects 0.000 description 1
- 229960002685 biotin Drugs 0.000 description 1
- 235000020958 biotin Nutrition 0.000 description 1
- 239000011616 biotin Substances 0.000 description 1
- 210000000601 blood cell Anatomy 0.000 description 1
- 238000010241 blood sampling Methods 0.000 description 1
- 230000004579 body weight change Effects 0.000 description 1
- 210000000988 bone and bone Anatomy 0.000 description 1
- 210000002798 bone marrow cell Anatomy 0.000 description 1
- 210000004271 bone marrow stromal cell Anatomy 0.000 description 1
- 210000004958 brain cell Anatomy 0.000 description 1
- 201000011510 cancer Diseases 0.000 description 1
- 230000036952 cancer formation Effects 0.000 description 1
- 150000001720 carbohydrates Chemical class 0.000 description 1
- 235000014633 carbohydrates Nutrition 0.000 description 1
- 231100000504 carcinogenesis Toxicity 0.000 description 1
- 230000000747 cardiac effect Effects 0.000 description 1
- 230000021164 cell adhesion Effects 0.000 description 1
- 230000022131 cell cycle Effects 0.000 description 1
- 230000030833 cell death Effects 0.000 description 1
- 230000010261 cell growth Effects 0.000 description 1
- 238000002659 cell therapy Methods 0.000 description 1
- 210000004978 chinese hamster ovary cell Anatomy 0.000 description 1
- 230000009816 chondrogenic differentiation Effects 0.000 description 1
- 210000003483 chromatin Anatomy 0.000 description 1
- 210000001072 colon Anatomy 0.000 description 1
- 230000005757 colony formation Effects 0.000 description 1
- 230000000295 complement effect Effects 0.000 description 1
- 238000010276 construction Methods 0.000 description 1
- 238000011109 contamination Methods 0.000 description 1
- 238000007796 conventional method Methods 0.000 description 1
- 210000000399 corneal endothelial cell Anatomy 0.000 description 1
- 239000006059 cover glass Substances 0.000 description 1
- 238000005138 cryopreservation Methods 0.000 description 1
- 239000012228 culture supernatant Substances 0.000 description 1
- 230000006378 damage Effects 0.000 description 1
- 238000007405 data analysis Methods 0.000 description 1
- 230000002950 deficient Effects 0.000 description 1
- 210000005258 dental pulp stem cell Anatomy 0.000 description 1
- 230000030609 dephosphorylation Effects 0.000 description 1
- 238000011161 development Methods 0.000 description 1
- 230000018109 developmental process Effects 0.000 description 1
- 229960003957 dexamethasone Drugs 0.000 description 1
- UREBDLICKHMUKA-CXSFZGCWSA-N dexamethasone Chemical compound C1CC2=CC(=O)C=C[C@]2(C)[C@]2(F)[C@@H]1[C@@H]1C[C@@H](C)[C@@](C(=O)CO)(O)[C@@]1(C)C[C@@H]2O UREBDLICKHMUKA-CXSFZGCWSA-N 0.000 description 1
- 235000014113 dietary fatty acids Nutrition 0.000 description 1
- BNIILDVGGAEEIG-UHFFFAOYSA-L disodium hydrogen phosphate Chemical compound [Na+].[Na+].OP([O-])([O-])=O BNIILDVGGAEEIG-UHFFFAOYSA-L 0.000 description 1
- 229940079593 drug Drugs 0.000 description 1
- 238000004520 electroporation Methods 0.000 description 1
- 239000008393 encapsulating agent Substances 0.000 description 1
- 238000005516 engineering process Methods 0.000 description 1
- 108010048367 enhanced green fluorescent protein Proteins 0.000 description 1
- 210000001339 epidermal cell Anatomy 0.000 description 1
- 210000002514 epidermal stem cell Anatomy 0.000 description 1
- 210000003560 epithelium corneal Anatomy 0.000 description 1
- 210000002744 extracellular matrix Anatomy 0.000 description 1
- 239000000194 fatty acid Substances 0.000 description 1
- 229930195729 fatty acid Natural products 0.000 description 1
- 150000004665 fatty acids Chemical class 0.000 description 1
- 238000001914 filtration Methods 0.000 description 1
- MHMNJMPURVTYEJ-UHFFFAOYSA-N fluorescein-5-isothiocyanate Chemical compound O1C(=O)C2=CC(N=C=S)=CC=C2C21C1=CC=C(O)C=C1OC1=CC(O)=CC=C21 MHMNJMPURVTYEJ-UHFFFAOYSA-N 0.000 description 1
- 238000002073 fluorescence micrograph Methods 0.000 description 1
- 239000012737 fresh medium Substances 0.000 description 1
- 108020001507 fusion proteins Proteins 0.000 description 1
- 102000037865 fusion proteins Human genes 0.000 description 1
- 229920000159 gelatin Polymers 0.000 description 1
- 239000008273 gelatin Substances 0.000 description 1
- 235000019322 gelatine Nutrition 0.000 description 1
- 235000011852 gelatine desserts Nutrition 0.000 description 1
- 230000002068 genetic effect Effects 0.000 description 1
- 210000004602 germ cell Anatomy 0.000 description 1
- 239000003102 growth factor Substances 0.000 description 1
- 230000007773 growth pattern Effects 0.000 description 1
- 231100001261 hazardous Toxicity 0.000 description 1
- 229960002897 heparin Drugs 0.000 description 1
- 229920000669 heparin Polymers 0.000 description 1
- 229940088597 hormone Drugs 0.000 description 1
- 239000005556 hormone Substances 0.000 description 1
- 229960000890 hydrocortisone Drugs 0.000 description 1
- 238000010191 image analysis Methods 0.000 description 1
- 210000000987 immune system Anatomy 0.000 description 1
- 230000001976 improved effect Effects 0.000 description 1
- 238000000338 in vitro Methods 0.000 description 1
- 230000002779 inactivation Effects 0.000 description 1
- 229960000905 indomethacin Drugs 0.000 description 1
- 230000000977 initiatory effect Effects 0.000 description 1
- 229940047122 interleukins Drugs 0.000 description 1
- 230000000968 intestinal effect Effects 0.000 description 1
- 230000003834 intracellular effect Effects 0.000 description 1
- 238000001638 lipofection Methods 0.000 description 1
- 238000011068 loading method Methods 0.000 description 1
- 210000005265 lung cell Anatomy 0.000 description 1
- 238000002826 magnetic-activated cell sorting Methods 0.000 description 1
- 239000003550 marker Substances 0.000 description 1
- 238000005259 measurement Methods 0.000 description 1
- 230000031864 metaphase Effects 0.000 description 1
- 108091070501 miRNA Proteins 0.000 description 1
- 239000002679 microRNA Substances 0.000 description 1
- 239000011325 microbead Substances 0.000 description 1
- 238000000520 microinjection Methods 0.000 description 1
- 239000007758 minimum essential medium Substances 0.000 description 1
- 238000002156 mixing Methods 0.000 description 1
- 238000012986 modification Methods 0.000 description 1
- 230000004048 modification Effects 0.000 description 1
- 238000012544 monitoring process Methods 0.000 description 1
- 230000000877 morphologic effect Effects 0.000 description 1
- 230000008722 morphological abnormality Effects 0.000 description 1
- 210000000663 muscle cell Anatomy 0.000 description 1
- 108700024542 myc Genes Proteins 0.000 description 1
- 210000003098 myoblast Anatomy 0.000 description 1
- 210000002241 neurite Anatomy 0.000 description 1
- 230000004031 neuronal differentiation Effects 0.000 description 1
- 231100001083 no cytotoxicity Toxicity 0.000 description 1
- 210000004940 nucleus Anatomy 0.000 description 1
- 210000000287 oocyte Anatomy 0.000 description 1
- INAAIJLSXJJHOZ-UHFFFAOYSA-N pibenzimol Chemical compound C1CN(C)CCN1C1=CC=C(N=C(N2)C=3C=C4NC(=NC4=CC=3)C=3C=CC(O)=CC=3)C2=C1 INAAIJLSXJJHOZ-UHFFFAOYSA-N 0.000 description 1
- 210000002826 placenta Anatomy 0.000 description 1
- 210000001778 pluripotent stem cell Anatomy 0.000 description 1
- 229920000729 poly(L-lysine) polymer Polymers 0.000 description 1
- 239000002243 precursor Substances 0.000 description 1
- 102000004196 processed proteins & peptides Human genes 0.000 description 1
- 108090000765 processed proteins & peptides Proteins 0.000 description 1
- 239000000186 progesterone Substances 0.000 description 1
- 229960003387 progesterone Drugs 0.000 description 1
- 230000001737 promoting effect Effects 0.000 description 1
- 230000001902 propagating effect Effects 0.000 description 1
- 238000004445 quantitative analysis Methods 0.000 description 1
- 108700042226 ras Genes Proteins 0.000 description 1
- 210000003370 receptor cell Anatomy 0.000 description 1
- 210000000664 rectum Anatomy 0.000 description 1
- 230000009467 reduction Effects 0.000 description 1
- 230000022532 regulation of transcription, DNA-dependent Effects 0.000 description 1
- 230000003716 rejuvenation Effects 0.000 description 1
- 238000007634 remodeling Methods 0.000 description 1
- 238000010839 reverse transcription Methods 0.000 description 1
- 150000003839 salts Chemical class 0.000 description 1
- 229940082569 selenite Drugs 0.000 description 1
- MCAHWIHFGHIESP-UHFFFAOYSA-L selenite(2-) Chemical compound [O-][Se]([O-])=O MCAHWIHFGHIESP-UHFFFAOYSA-L 0.000 description 1
- 230000035945 sensitivity Effects 0.000 description 1
- 210000003491 skin Anatomy 0.000 description 1
- 210000004927 skin cell Anatomy 0.000 description 1
- 210000004988 splenocyte Anatomy 0.000 description 1
- 239000000126 substance Substances 0.000 description 1
- 230000004083 survival effect Effects 0.000 description 1
- 239000008399 tap water Substances 0.000 description 1
- 235000020679 tap water Nutrition 0.000 description 1
- 238000010257 thawing Methods 0.000 description 1
- 230000008719 thickening Effects 0.000 description 1
- 210000004746 tooth root Anatomy 0.000 description 1
- 230000002103 transcriptional effect Effects 0.000 description 1
- 239000012096 transfection reagent Substances 0.000 description 1
- 241000701161 unidentified adenovirus Species 0.000 description 1
- 210000003934 vacuole Anatomy 0.000 description 1
- 238000012795 verification Methods 0.000 description 1
- 230000029812 viral genome replication Effects 0.000 description 1
- 244000000009 viral human pathogen Species 0.000 description 1
- 230000009385 viral infection Effects 0.000 description 1
- 230000003612 virological effect Effects 0.000 description 1
- 239000011782 vitamin Substances 0.000 description 1
- 229940088594 vitamin Drugs 0.000 description 1
- 229930003231 vitamin Natural products 0.000 description 1
- 235000013343 vitamin Nutrition 0.000 description 1
- 238000005406 washing Methods 0.000 description 1
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 1
- 230000004580 weight loss Effects 0.000 description 1
Images
Classifications
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N5/00—Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
- C12N5/10—Cells modified by introduction of foreign genetic material
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N5/00—Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
- C12N5/06—Animal cells or tissues; Human cells or tissues
- C12N5/0602—Vertebrate cells
- C12N5/0652—Cells of skeletal and connective tissues; Mesenchyme
- C12N5/0662—Stem cells
- C12N5/0665—Blood-borne mesenchymal stem cells, e.g. from umbilical cord blood
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N5/00—Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
- C12N5/06—Animal cells or tissues; Human cells or tissues
- C12N5/0602—Vertebrate cells
- C12N5/0652—Cells of skeletal and connective tissues; Mesenchyme
- C12N5/0662—Stem cells
- C12N5/0663—Bone marrow mesenchymal stem cells (BM-MSC)
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/63—Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
- C12N15/79—Vectors or expression systems specially adapted for eukaryotic hosts
- C12N15/85—Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
- C12N15/86—Viral vectors
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2510/00—Genetically modified cells
- C12N2510/04—Immortalised cells
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2760/00—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
- C12N2760/00011—Details
- C12N2760/18011—Paramyxoviridae
- C12N2760/18811—Sendai virus
- C12N2760/18841—Use of virus, viral particle or viral elements as a vector
- C12N2760/18843—Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
Definitions
- the present invention relates to a method for producing a reversible immortalized cell, and more particularly to a method for producing a reversible immortalized cell using a chromosomal non-integrating RNA viral vector carrying a predetermined immortalizing gene.
- the main cell sources used for regenerative medicine and cell therapy are somatic stem cells such as mesenchymal stem cells (MSC) and pluripotent stem cells such as ES cells and iPS cells.
- MSC mesenchymal stem cells
- ES cells pluripotent stem cells
- iPS cells pluripotent stem cells
- stem cells mesenchymal stem cells (MSCs) are cells that originally exist in the body, so there is little risk of rejection and tumorigenesis, and they are excellent in terms of safety, but because the number of cell divisions is limited. The challenge is to secure a sufficient number of cells to supply to the medical field.
- telomerase reverse transcriptase genes that regulate the expression or activity of telomerase (for example, Myc gene, Ras gene, etc.), and viral genes (SV40T, HPV E6-E7, EBV, etc.) are known.
- vectors such as plasmid DNA, lentiviral vector, retroviral vector, and adenoviral vector are used for introduction of these immortalizing genes into cells (Patent Document 1, Patent Document 2, Patent Document 3, etc.).
- the virus vector is preferable in that the gene can be efficiently introduced by simply filtering the culture supernatant of the recombinant virus vector-producing cell and adding it to the target cell.
- the plasmid DNA and the adenovirus vector have a vector genome of DNA and are integrated into the host chromosome.
- the vector genome is RNA, but in the cell, the DNA is taken in the form of DNA by a reverse transcription enzyme and integrated into the chromosome of the host to express the loaded gene.
- the vector generally used for immortalization has a risk of damaging the gene of the host cell by incorporating foreign DNA into the chromosome of the host cell and, in some cases, forming a tumor, and thus the safety of regenerative medicine.
- the Sendai virus vector (SeV vector) is a non-chromosomally integrated RNA virus vector that does not take the form of DNA.
- the SeV vector can be introduced into a wide range of cell types and easily expresses proteins, so that it is widely used for inducing iPS cells (Patent Document 4).
- stem cells obtained from human tissues are a group of young cells and aged cells, and have various properties. They divide and age, so cells can be maintained at a constant quality. not. Therefore, there is a different problem that it is difficult to set quality standards, which are necessary elements for automation and mechanization.
- iPS cells Attempts to isolate only cells with the desired properties from those different cell populations are beginning to be discussed.
- Known methods for evaluating the quality of iPS cells include miRNA analysis, surface marker detection, cell morphology image analysis, and epigenome analysis, and MSC has also proposed the use of these techniques, but unlike iPS cells. Since MSCs do not grow indefinitely and cannot be cloned, they have not been put into practical use.
- the present invention is a method for producing a reversible immortalized cell, which can proliferate the cell for a long period of time without damaging the chromosome of the cell into which the immortalizing gene has been introduced, and can remove the immortalizing gene. It is an object of the present invention to provide a method for obtaining a large amount of reversible immortalized cells that can be cloned and have stable quality.
- the present inventors have obtained one or more immortalizing genes selected from the group consisting of Bmi-1 gene, TERT gene, and SV40T gene.
- MSC mesenchymal stem cells
- Sendai virus vector which is a non-chromosomal RNA vector that has not been used for cell immortalization
- it has been used for more than 80 days. It was found that cell proliferation does not occur and that infinite cell proliferation (immortalization) is possible, and that the rate of cell proliferation increases at an early stage after gene transfer. It was also confirmed that the produced immortalized cells had no chromosomal abnormality, had pluripotency, and could easily remove the immortalized gene by temperature control.
- the immortalized cells were successfully cloned from the obtained immortalized cell population, and 8 out of 10 clones of the cloned immortalized cells also showed normal nuclear type and were pluripotent. It was confirmed that immortalization can be induced in cells at any stage from young cells to senescent cells in which division has stopped, and that cloning is possible immediately after immortalization. The present invention has been completed based on such findings.
- the present invention includes the following inventions.
- a method for producing a reversible immortalized cell which comprises the following steps. (1) A step of introducing a chromosome-non-integrating RNA viral vector carrying an immortalizing gene into mammalian cells and expressing the immortalizing gene in the cells, and (2) culturing the cells obtained in step (1). And the process of growing [2] The method according to [1], wherein the immortalizing gene is one or more immortalizing genes selected from the group consisting of the Bmi-1 gene, the TERT gene, and the SV40T gene. [3] The method according to [1] or [2], wherein the immortalizing gene is any of the following (a) to (d).
- RNA virus vector is a paramyxovirus vector.
- paramyxovirus vector is a Sendai virus vector.
- Sendai virus vector is a temperature sensitive Sendai virus vector.
- chromosomal non-integrating RNA virus vector is a Sendai virus vector, further comprising a step of removing the Sendai virus vector after culturing in the step (2).
- removal of the Sendai virus vector is performed by changing the culture temperature from 35 ° C to 37 ° C.
- a cell that is difficult to mass-culture for example, a mesenchymal stem cell (MSC) used for regenerative medicine
- MSC mesenchymal stem cell
- a temperature-sensitive Sendai virus vector (SeV vector) as a non-chromosomal integrated RNA vector, the vector can be easily removed from cells only by changing the culture temperature without performing a complicated removal operation. Therefore, it is possible to supply a large amount of cells having excellent safety.
- the immortalized cells obtained by the present invention can be used for regenerative medicine because they have no chromosomal abnormality and have pluripotency as before gene transfer.
- it is difficult to clone non-immortalized cells because they have different lifespans and properties, but the immortalized cells produced by the present invention are easy to clone and can stably obtain high-quality cells. Therefore, the mechanization of cell manufacturing facilitates quality control, makes it possible to significantly reduce the manufacturing cost, which is an issue for industrialization, and contributes to the development of cell medicine and regenerative medicine.
- the genomic structure of the SeV vector carrying the immortalizing factor gene is shown [A. SeV (18+) Bmi1 (HNL) OFP / TS15d ⁇ F, B. SeV (PM) hTERT (HNL) EGFP / TS15 ⁇ F, C. SeV (18+). ) SV40T / TS15 ⁇ F, D. SeV (HNL) E6-E7, BFP / TS15 ⁇ F].
- Transmitted light and fluorescence (OFP) images of Bmi-1 solitary infected cells (MOI: 1,5,20), transmitted light and fluorescence (GFP) images of hTERT solitary infected cells (MOI: 1,5,20) show.
- the transmitted light image and the fluorescence (OFP, GFP) image of the two-factor (Bmi-1 / hTERT) co-infected cell (MOI: 1,5,20) are shown.
- the transmitted light image and the fluorescence (OFP, GFP) image of the three-factor (Bmi-1 / hTERT / SV40T) co-infected cell (MOI: 1,5,20) are shown.
- the cell morphology and cell condition determination 59 days after the introduction of immortalizing factors (No. 1 to No. 16) are shown. Shows the cell proliferation curve of immortalizing factors (No. 1, No. 2, No. 4, No. 12, No. 16) introduced cells (up arrow: morphological (transmitted light) image and fluorescent image point (Fig. 5)).
- the transmitted light image and the fluorescence (OFP, GFP) image of the three-factor (Bmi-1 / hTERT / SV40T) -introduced cell are shown.
- the morphology (transmitted light) image of the three-factor (Bmi-1 / hTERT / SV40T) -introduced cell is shown (maintained at 35 ° C, temperature changed from 35 ° C to 37 ° C).
- the chromosomal analysis results (normal karyotype) after 90 days of subculture of the three-factor (Bmi-1 / hTERT / SV40T) -introduced cells are shown (chromosome analysis point ( ⁇ ) in FIG. 7).
- telomere analysis points (A) The results of quantitative analysis of telomeres in long-term culture samples of immortalizing factors (No.1, No.2, No.4, No.12, No.16) are shown (A. Effects of immortalizing factor combinations on telomeres). (Day0, Day40: Telomere analysis point in Fig. 6 (*)), B. After introduction of factor 3 (Bmi-1 / hTERT / SV40T), change in telomere amount due to factor non-removal / removal (Day0, Day40: Fig. 6) Telomere analysis points (*), Day85, Day98: Telomere analysis points in Fig. 7 (*)). Pictures of adipocytes differentiated from early cultured MSCs and immortalized MSCs are shown.
- clone A10 The results of chromosomal analysis of cloned immortalized cells (clone A10) are shown (Day 80: Chromosome analysis point ( ⁇ ) in FIG. 18). It shows the differentiation potential of cloned immortalized cells (fat cell differentiation, neuronal cell differentiation, osteoblast differentiation).
- Cryopreservation points of MSCs used in single-cell cloning studies (early MSCs: Day9, mid-term MSCs: Day24, late MSCs: Day49) and immortalization induction points by SeV vector infection (early MSCs: Day21, mid-term MSCs: Day36, late MSCs) : Day61) is shown. Photographs of SeV vector infected cells (early MSCs, middle MSCs, late MSCs) and SeV vector non-infected MSCs (controls) before single-cell cloning (immediately before 96-well plate seeding) are shown.
- results of a single cell cloning test of cell populations of SeV vector non-infected MSC (control) and SeV vector infected cells (early MSC) are shown (numbers in wells: number of clones formed, culture days: 2 weeks).
- results of a single cell cloning test of a cell population of SeV vector-infected cells are shown (numbers in wells: number of clones formed, number of culture days: 2 weeks).
- Photographs of SeV vector infection test results for cells in which cell division has stopped Day 90 (cells immediately before SeV vector infection, SeV vector non-infected cells (control cells), and SeV vector infected cells (2 weeks after infection)) ).
- the test results of single-cell cloning of arrested cells (Day 90) that started reproliferation by SeV vector infection are shown (numbers in wells: number of clones formed, number of culture days: 2 weeks).
- the cell proliferation curve of adipose tissue-derived human MSC (non-infected cell line, SeV vector infected cell line) in serum-free culture is shown.
- the cell proliferation curve of bone marrow-derived human MSC (non-infected cell line, SeV vector infected cell line) in serum-free culture is shown.
- a transmitted light image and a fluorescence (OFP, GFP) image of a SeV vector-infected cell line of a serum-free cultured adipose tissue-derived human MSC (hMSC-AT) and a SeV vector-infected cell line of a bone marrow-derived human MSC (hMSC-BM) are shown.
- hMSC-AT 16th and 58th days from the date of infection
- hMSC-BM 24th and 50th days from the date of infection
- the cell proliferation curve of rat MSC non-infected cell line, SeV vector infected cell line # 1, # 2 is shown.
- Transmission light images and fluorescence (OFP, GFP) images of SeV vector-infected cell lines of immortalized rat MSC are shown (2nd, 9th, and 58th days from the day of infection).
- the cell proliferation curve of HFL1 non-infected cell line # 1, # 2, SeV vector infected cell line # 1, # 2, 37 ° C cultured SeV vector infected cell line # 1, # 2) is shown.
- HUVEC non-infected cell line # 1, SeV vector infected cell line # 1, # 2 cultured at 35 ° C, SeV vector infected cell line # 1, # 2 cultured at 37 ° C
- Immortalized HUVEC [35 ° C culture (7 days, 36, 72 days from infection date), 35 ° C culture + 37 ° C culture (35 days + 37 days from infection date)] transmitted light images and fluorescence (OFP, GFP) images are shown. .. It is a photograph showing the result of FISH analysis of immortalized MSC by introduction of a multiproliferative factor-inserted human artificial chromosome vector (in the lower right frame: DAPI-stained image of arrow cells). The results of a healing effect test of an enteritis model by transplantation of immortalized MSCs are shown.
- the present invention is a method for producing reversible immortalized cells, wherein the method (1) introduces a chromosomal non-integrating RNA virus vector carrying an immortalizing gene into mammalian cells. Then, the step of expressing the immortalizing gene in the cell and the step of (2) culturing and proliferating the cell obtained in the step (1) are included.
- the "cell” refers to all somatic cells other than germline cells (egg and sperm, oocyte, ES cell, etc.) and totipotent cells (iPS cells).
- the "somatic cell” may be any of a primary cultured cell, a subcultured cell, and an established cell line.
- the somatic cells may be naturally derived or artificially produced by differentiating from iPS cells or the like. Specific examples of somatic cells include cells that form tissues (fat cells, fibroblasts, nerve cells, skin cells, blood cells, muscle cells, osteoblasts, cartilage cells, hepatocytes, pancreatic cells, and renal cells.
- Myocardial cells brain cells, lung cells, splenocytes, adrenal cells, gingival cells, root membrane cells) or their precursor cells, immune system cells (B cells, T cells, monocytic cells) (Cells, etc.), somatic stem cells [mesophageal stem cells (adipose-derived stem cells, bone marrow-derived stem cells, umbilical cord blood-derived stem cells, placenta-derived stem cells, etc.), hematopoietic stem cells, nerve stem cells, epidermal stem cells, intestinal epithelial stem cells, dental pulp stem cells, tooth roots, etc. Membrane stem cells, etc.] and the like.
- the origin of the cells is not particularly limited as long as it is a mammal, and examples thereof include humans, mice, rats, guinea pigs, hamsters, rabbits, dogs, cats, pigs, cows, and horses.
- the "immortalized cell” refers to a cell whose proliferation does not stop even if cell division is repeated, that is, a cell having an infinite proliferation ability, unlike a primary cultured cell or a cell cultured under normal culture conditions.
- the "immortalized cell” in the present invention means a cell capable of infinite proliferation by introducing a predetermined immortalizing gene, and a cell whose infinite proliferation ability does not decrease even if it is repeatedly subcultured and cultured. ..
- the “immortalized cells” according to the present invention have different growth rates and growth periods depending on the origin of the cells or the culture conditions, but as a result of subculture, untreated cells proliferate or cease under the same culture conditions.
- the immortalized cells of the present invention include both the cell population capable of infinite proliferation as described above and the immortalized cell line cloned from the cell population.
- reversible immortalization means that a cell proliferation is stopped or attenuated by introducing an immortalizing gene into a cell, making the cell proliferative indefinitely, and then removing the immortalizing gene. It means to let.
- “Immortalization” means to release the limit of the number of cell divisions and cell aging of early cells, and to impart continuous cell division ability and proliferation ability. Specifically, under standard cell culture conditions, usually 5 or more passages, preferably 7 or more passages, 8 or more passages, 9 or more passages, 10 or more passages, 12 or more passages, 15 passages or more. As mentioned above, it means that more than 20 passages have become possible.
- Cells having a confluent number of passages of 0 can be amplified and cultured by subculturing using a method known to those skilled in the art. The cells obtained by one passage operation are called “passage number 1 (or second generation)" cells, and “passage number 2, 3, 4 " corresponding to the number of passage operations. It can be expressed as "n (n (integer) is the number of passages) (n + 1th generation)".
- a step of freezing the cells may be included between each passage operation.
- the immortalized cell of the present invention is produced by introducing a predetermined immortalizing gene into a cell using a chromosome non-integrating RNA viral vector.
- the "immortalizing gene” refers to a gene that immortalizes a cell to acquire infinite proliferation ability and does not induce cell death.
- the immortalizing gene is an extrinsic gene and means an immortalizing gene newly introduced from the outside of the cell.
- the immortalizing gene may be an immortalizing gene derived from other than human, or may be an immortalizing gene modified into a form expressible in a target cell.
- the immortalizing gene one or more genes selected from the group consisting of the Bmi-1 gene, the TERT gene, and the SV40T gene can be used, but a combination of the two genes can be used. Preferably, a combination of three genes is more preferred.
- the TERT gene is preferable, and examples of a preferable combination when two or more kinds of genes are combined include a combination of Bmi-1 gene and TERT gene, and a combination of TERT gene and SV40T gene.
- the "gene” includes not only a structural gene that defines the primary structure of a protein but also a region on a nucleic acid having a control function such as a promoter or an operator. Therefore, in the present invention, the term “gene” is used without distinguishing between a regulatory region, a coding region, an exon, and an intron, unless otherwise specified.
- Bmi-1 (B lymphoma Mo-MLV insertion region 1 homolog) gene
- PRC1 polycomb repressive complex 1
- Bmi-1 is involved in the regulation of expression of various genes including Hox gene by controlling chromatin remodeling and histone modification.
- Bmi-1 has the effect of controlling cell proliferation by suppressing the expression of p16 and p19Arf involved in the cell cycle, and plays an important role in maintaining self-renewal by being involved in cell division of hematopoietic stem cells and nerve stem cells. It is known to play.
- TERT telomerase reverse transcriptase gene
- TERT Telomerase reverse transcriptase gene
- Telomerase RNA component extends specific repeat sequences of eukaryotic chromosome ends (telomeres) from telomere RNA components (Telomerase RNA: TR or Telomerase RNA component: TERC) and other regulatory subunits. It constitutes the enzyme telomerase. It is known that TERT has a role in maintaining telomere length, whereas cell aging has a telomere length monitoring mechanism and cell senescence is caused by telomere shortening.
- the "SV40T (simianvirus 40large Tantigen) gene” is a gene encoding the Simian virus 40 large T antigen.
- the genome of SV40 (simianvacuolating virus40) is divided into an early region that is expressed immediately after infection, a late region that is expressed during replication of the viral genome after infection, and a regulatory region that includes transcriptional regulation and the origin of replication.
- the early region encodes a large T antigen involved in the initiation of viral genome replication and inactivation of p53 and pRB, which are tumor suppressor gene products, and a small t antigen that binds to and inhibits the protein dephosphorylation enzyme PP2A.
- a specific example of the Bmi-1 gene used in the present invention is the mouse BMI1 gene (SEQ ID NO: 1)
- a specific example of the TERT gene is the human TERT gene (SEQ ID NO: 2)
- a specific example of the SV40T gene is SV40 large.
- the T antigen gene (SEQ ID NO: 3) can be mentioned.
- the Bmi-1 gene, TERT gene, and SV40T gene may also be these transcriptional variants, splicing variants and their orthologs.
- the above Bmi-1 gene, TERT gene, and SV40T gene are 80% or more, preferably 90% or more, relative to each of the nucleotide sequences of SEQ ID NOs: 1, 2, and 3 as long as they have the same functions and activities.
- the Bmi-1 gene, TERT gene, and SV40T gene are artificially modified so that the product of the gene is expressed as a fusion protein with other proteins or peptides as long as it has the same function and activity. It may be a gene.
- a "chromosome non-integrating RNA virus vector” is used as a vector for introducing and expressing the immortalization gene in cells.
- the viral vector means a vector having a genomic nucleic acid derived from the virus and capable of expressing the gene by incorporating a transgene into the nucleic acid.
- the "chromosome non-integrated RNA virus vector” is a virus vector derived from a virus and capable of introducing a gene into a target cell, and the introduced gene is integrated into a host chromosome (nuclear-derived chromosome). A non-hazardous carrier.
- RNA virus vector refers to a vector consisting of a virus containing RNA of minus strand (antisense strand complementary to the sense strand encoding the viral protein) as a genome, and minus strand RNA is also called negative strand RNA. ..
- a single-stranded negative-strand RNA virus also referred to as a non-segmented negative-strand RNA virus is particularly preferable.
- Single-stranded negative-strand RNA virus refers to a virus having a single-stranded negative-strand (minus-strand) RNA in its genome, for example, Paramyxoviridae; Paramyxovirus, Morbillivirus, Rubulavirus, Pneumovirus, etc. Includes), Rhabdoviridae (including Rhabdoviridae; Vesculovirus, Lyssavirus, and Ephemerovirus genus), and viruses belonging to the family Filoviridae.
- minus-strand RNA viruses examples include Paramyxoviridae virus Sendai virus, Newcastle disease virus, Mumps virus, and measles virus.
- RS virus Respiratory syncytial virus
- cattle epidemic virus Rinderpest virus
- distemper virus distemper virus
- salparainfluenza virus SV5
- human parainfluenza virus type 1,2,3, orthomixovirus family Orthomyxoviridae influenza virus (Influenza virus), Rhabdoviridae (Rhabdoviridae) vesicular stomatitis virus (Vesicular stomatitis virus), mad dog disease virus (Rabies virus) and the like can be mentioned, but Sendai virus (Sendai virus) is preferable.
- Sendai virus vector SeV vector
- a Sendai virus (SeV) vector is used as the non-chromosomal integrated RNA vector.
- the immortalizing factor genes of the Bmi-1 gene, TERT gene, and SV40T gene may be inserted separately into the SeV vector, or may be inserted together into a single SeV vector.
- Sendai virus is a type of virus belonging to the genus Respirovirus of the Paramyxoviridae family, and contains a single minus strand (antisense strand against the sense strand encoding the viral protein) RNA as a genome.
- the Sendai virus genome consists of NP (nucleocapsid) gene, P (phospho) gene, M (matrix) gene, F (fusion) gene, and HN (hemogglutinin / neuraminidase) in order from the 3'end to the 5'end. ) Gene and L (large) gene are included. Of these, Sendai virus can sufficiently function as a vector if it has the NP gene, P gene, and L gene, and can replicate the genome in cells and express the loaded gene. Since the Sendai virus has minus-strand RNA in its genome, the 3'side of the genome is upstream and the 5'side is downstream, which is the opposite of the normal case.
- the SeV vector can be used as a natural strain, a wild strain, a mutant strain, or a commercially available product.
- the virus may be a virus having a structure similar to that of a virus isolated from nature, or a virus artificially modified by genetic recombination, as long as the desired function can be achieved.
- any gene possessed by the wild-type virus may be mutated or deleted.
- a non-propagating vector ( ⁇ F) in which the F gene is deleted from the genome and no infectious particles are formed from the gene-introduced cells, and the F gene is deleted, and the M and / or HN gene is deleted.
- a vector that further deletes or has a mutation in the M and / or HN gene is preferably used in the present invention.
- a vector in which the F gene is deleted, the M or HN gene is further deleted, and the remaining M and / or HN gene is further mutated is also preferably used in the present invention (patented). See No. 5763340, etc.).
- the SeV vector used in the method of the present invention is preferably temperature sensitive.
- “Temperature sensitivity” means that the activity is significantly reduced at a normal cell culture temperature (for example, 37 to 38 ° C) as compared with a low temperature (for example, 30 to 36 ° C).
- Sendai virus TS7 L protein Y942H / L1361C / L1558I mutation
- TS12 P protein D433A / R434A / K437A mutation
- TS13 P protein D433A / R434A / K437A mutation and L protein L1558I mutation
- TS14 P.
- Mutations such as protein D433A / R434A / K437A mutations and L protein L1361C) and TS15 are temperature-sensitive mutations and are preferred in the present invention. These mutations can be utilized. It is preferable to further introduce these mutations into the above-mentioned F gene-deficient SeV vector.
- SeV vectors refer to Japanese Patent No. 5763340, WO2015 / 046229, etc. be able to.
- the SeV vector in the present invention is a complex consisting of an infectious virus particle, a virus core, a complex of a virus genome and a virus protein, a non-infectious virus particle, and the like, and is loaded by introduction into cells. Includes a complex capable of expressing a gene that does.
- a ribonucleoprotein core part of a virus consisting of a Sendai virus genome and Sendai virus proteins (NP, P, and L proteins) that bind to it expresses the introduced gene in the cell by introducing it into the cell.
- the introduction into cells may be carried out by appropriately using a transfection reagent or the like. Therefore, such a ribonucleoprotein (RNP) is also included in the SeV vector in the present invention.
- the position where the immortalizing gene (Bmi-1 gene, TERT gene, and SV40T gene) is integrated is not particularly limited, but when each immortalizing gene is inserted into multiple separate vectors, the Bmi-1 gene is the NP gene. Upstream, the TERT gene is preferably inserted between the P and M genes, and the SV40T gene is preferably inserted upstream of the NP gene. Also, two or more genes (Bmi-1 and TERT, TERT and SV40T, or Bmi-1 and TERT and SV40T) may be inserted into a single vector.
- the SeV vector carrying the immortalization gene obtained as described above is introduced into the cells by adding the vector (Sendai virus particles) to the medium of the somatic cells and infecting the cells with the virus. .. Since the dose of the vector varies depending on the cell type, cell density, and amount of medium, the MOI whose infection efficiency is close to 100% may be investigated and determined in advance for each cell to be used.
- the SeV vector when in the form of RNP, it can be introduced into cells by a method such as an electroporation method, a lipofection method, or a microinjection method.
- the method for culturing cells into which an immortalizing gene has been introduced can be carried out according to the method and conditions for culturing normal mammalian somatic cells.
- the medium used for culturing is not particularly limited, and a medium generally used for maintenance culture or expansion culture of cells and suitable for virus infection may be used, and either a commercially available medium or a self-made medium may be used. good.
- a basal medium containing components necessary for cell survival and proliferation inorganic salts, carbohydrates, hormones, essential amino acids, non-essential amino acids, vitamins, fatty acids
- D-MEM Dulbecco's Modified Eagle's Medium
- Dulbecco's Modified Eagle's Medium Natural Mixture F-12 (D-MEM / F-12) Medium, Glassgow MEM (G-MEM) Medium, Basal Medium Eagle (BME) Medium, Minimum Essential Medium (MEM) Medium, Eagle's minimal essential Examples include medium (EMEM) medium, Iscove's Modified Dulbecco's Medium (IMDM) medium, RPMI 1640 medium, Medium 199 medium, ⁇ MEM medium, ham medium, Fischer medium, and a mixed medium thereof.
- D-MEM / F-12 Natural Mixture F-12
- G-MEM Basal Medium Eagle
- MEM Minimum Essential Medium
- the medium may contain growth factors (FGF, EGF, etc.), interleukins, insulin, transferase, heparin, heparan sulfate, collagen, fibronectin, progesterone, selenite, B27-supplement, N2-supplement, antibiotics, if necessary. It may contain a substance (penicillin, streptomycin, etc.) and the like. Further, the medium may be a serum-containing medium or a serum-free medium. From the viewpoint of preventing contamination of heterologous animal-derived components, it is preferable to use serum that does not contain serum or that is derived from the same animal as the cells to be cultured. Alternatively, a serum substitute such as albumin may be used.
- the culture method is not limited, but is limited to three-dimensional culture under non-adhesive conditions, for example, suspension culture (for example, dispersion culture, aggregate suspension culture, etc.), or two-dimensional culture under adhesive conditions, for example, flat plate. Examples thereof include culture and culture in which three-dimensional culture and two-dimensional culture are combined.
- the incubator used for culturing cells is not particularly limited as long as it can cultivate cells, and examples thereof include flasks, petri dishes, dishes, plates, chamber slides, tubes, trays, culture bags, and roller bottles. ..
- the incubator may be cell non-adhesive or adhesive, and is appropriately selected depending on the intended purpose.
- cell adhesion incubator one treated with a cell-supporting substrate or the like using an extracellular matrix or the like may be used for the purpose of improving the adhesion to cells.
- the cell-supporting substrate include collagen, gelatin, poly-L-lysine, poly-D-lysine, laminin, fibronectin and the like.
- the culture temperature is 30 ° C to 36 ° C, preferably 32 ° C to 35 ° C, and more preferably 33 to 35 ° C. Culturing is carried out in an atmosphere of CO 2 containing air, for example, at a CO 2 concentration of 2% to 5%.
- the culture temperature for removing the immortalizing gene is 37 ° C to 38 ° C, preferably 37 ° C to 37.5 ° C.
- the immortalized cells prepared as described above can be induced to differentiate into specific tissue cells by culturing them in a differentiation-inducing medium.
- the immortalized cell when it is a mesenchymal stem cell, it can be differentiated into adipocyte, osteoblast, nerve cell, chondrocyte and the like.
- composition of the medium for inducing the differentiation of the immortalized cells according to the present invention into the target cells, the differentiation-inducing factor, the culture method, the passage method, etc. can be appropriately set from well-known and conventional techniques.
- the differentiation-inducing medium can be appropriately selected according to the type of cells targeted for differentiation-inducing. Differentiation-inducing media for various tissues (mediums to which at least one type of differentiation-inducing or promoting factor according to the target cell for differentiation-inducing is added) are commercially available, and these commercially available media may be used.
- the medium for inducing the differentiation of immortalized cells according to the present invention into adipocytes is a commercially available adipocyte-inducing medium or a commercially available animal cell medium containing insulin, dexamesazone, indomethacin, or 3-isobutyl-1-methylxanthin. , A medium containing troglycazone, biotin, etc. can be used.
- Examples of commercially available media include Mesenchymal Stem Cell Adipogenic Differentiation Medium 2 (manufactured by PromoCell), Human Mesenchymal Stem Cell Adipogenic Differentiation Medium BulletKit8 (manufactured by Lonza), and the like.
- the medium for inducing the differentiation of the cells according to the present invention into osteoblasts includes dexamesazone, ascorbic acid, ⁇ -glycerophosphate, hydrocortisone, BMP4, in a commercially available osteoblast-inducing medium or a commercially available animal cell medium.
- a medium containing BMP2 etc. can be used.
- Examples of commercially available media include Mesenchymal Stem Cell Osteogenic Differentiation Medium (manufactured by PromoCell), Human Mesenchymal Stem Cell Osteogenic Differentiation Medium Bullet Kit (manufactured by Lonza), and the like.
- a medium for inducing differentiation of immortalized cells into nerve cells according to the present invention, a commercially available nerve cell culture medium or nerve differentiation medium (for example, Mesenchymal Stem Cell Neurogenic Differentiation Medium (manufactured by PromoCell), etc.) can be used.
- the nerve cell culture medium or the nerve differentiation inducing medium preferably contains a nerve cell inducing factor (for example, brain-derived neurotrophic factor (BDNF), fibroblast growth factor (FGF)).
- BDNF brain-derived neurotrophic factor
- FGF fibroblast growth factor
- chondrocyte-inducing medium a commercially available chondrocyte-inducing medium or a commercially available animal cell medium containing dexamesazone, ascorbic acid, and TGF- ⁇ 3 is used.
- examples of commercially available media include MesenchymalStemCellChondrogenic DifferentiationMedium (manufactured by PromoCell), HumanMesenchymalStemCellChondrogenicDifferentiationMedium BulletKit (manufactured by Lonza), and the like.
- the culture conditions for inducing differentiation are the same as the culture conditions for culturing normal stem cells.
- the culture period for inducing differentiation is also not particularly limited, but is generally 5 to 20 days, preferably 7 to 18 days.
- stem cells have been induced to differentiate into the target cells can be confirmed by examining the expression of markers specific to each differentiated cell. For example, differentiation into adipocytes can be confirmed by oil red 0 staining, differentiation into osteoblasts by alkaline phosphatase staining, differentiation into nerve cells by NeuroFluorNeuO staining, and differentiation into chondrocytes by alcian blue staining.
- the immortalized cells and tissue cells induced to differentiate from immortalized (stem) cells obtained by the present invention can be used, for example, by transplanting cells to a diseased or damaged site, and can be provided as a product for regenerative medicine.
- products for regenerative medicine include cultured skin, cultured cartilage, cultured corneal epithelium, various cell sheets (for example, epidermal cell sheet, fibroblast sheet, corneal endothelial cell sheet, myocardial cell sheet, osteoblast sheet, myoblast).
- the serum-containing medium for culturing human mesenchymal stem cells is D-MEM (Low Gulucose), 20% FBS, 0.01 mol / L Hepes, Penicillin 100units / ml, Streptomycin 100 ⁇ g / It was prepared with a composition of ml and bFGF 20 ng / ml. Since bFGF has a very short half-life, Gibco Heat Stable Recombinant Human bFGF (Thermo Fisher Scientific), which has excellent stability, was used. MSCs were cultured in the above medium in a 5% CO 2 incubator. The human MSC used in the experiment is either bone marrow-derived, adipose tissue-derived, cord blood-derived, or umbilical cord matrix-derived.
- Example 1 Preparation of seV vector carrying an immortalizing gene and examination of infection conditions
- a SeV vector carrying an immortalizing gene Bmi-1 B lymphoma Mo-MLV insertion region 1 homolog
- Bmi-1 B lymphoma Mo-MLV insertion region 1 homolog
- HTERT human telomerase reverse transcriptase
- SV40T simian virus 40 large T antigen
- E6 / E7 human papillomavirus 16 E6 protein and E7 protein
- Bmi was used as the SeV vector for the non-chromosomal RNA virus vector.
- FIG. 1 shows the structure of each vector.
- Bmi-1, SV40T is the vector genome. It was installed at the most upstream of. The mounting positions of hTERT and E6 / E7 were changed downstream due to the decrease in vector production efficiency, which is thought to be due to high expression.
- SeV vector For the SeV vector, we used a TS15 ⁇ F type vector that was improved so that the viral vector disappeared from the cells by changing the culture temperature from 35 ° C to 37 ° C (Efficient generation of transgene-free human induced pluripotent stem cells).
- iPSCs by temperature-sensitive Sendai virus vectors. Ban H, Nishishita N, Fusaki N, Tabata T, Saeki K, Shikamura M, Takada N, Inoue M, Hasegawa M, Kawamata S, Nishikawa . 2011 Aug 23; 108 (34): 14234-9.).
- bone marrow-derived mesenchymal stem cells product name: ultra-high-purity human mesenchymal stem cells (REC), PuREC Co., Ltd.
- REC ultra-high-purity human mesenchymal stem cells
- PuREC Co., Ltd. are placed in a 48-well plate (FALCON 353230) supplemented with a medium of 200 ⁇ l / well in 5 ⁇ 10 4 Sprinkle individual / well (70-80% confluent) and add 4 types of immortalizing factor gene-laden SeV vectors to the cells so that the MOI of each vector becomes 1,5,20 by single or multiple co-infection. , Incubated overnight. The next day, the medium was replaced with fresh medium, and the medium was replaced every 2 days thereafter. The medium was expanded to a 12-well plate (FALCON 353043) on the 5th day after the vector infection, and the cells were observed 9 days after the infection.
- Example 2 Selection of immortalizing factor MSC by infecting MSC with a SeV vector carrying four kinds of immortalizing factor (Bmi-1, hTERT, SV40T, E6 / E7) genes alone and culturing for a long period of time. The factors necessary for immortalization of the disease were selected.
- immortalizing factor Bmi-1, hTERT, SV40T, E6 / E7
- MSC derived from umbilical cord blood (product name: Umbilical Cord-Derived Mesenchymal Stem Cells; Normal, Human (ATCC PCS-500-010)) was used for the study, and one or a combination of two or more SeV vectors carrying the immortalizing factor gene was used. (15 ways in total) were infected with the above MSC (48 well plate, 3 ⁇ 10 4 / well, MOI: 20). MSCs not infected with SeV vector were used as negative controls.
- the cells are expanded and cultured in the order of 48 well ⁇ 12 well ⁇ 6 well, and after that, when the cells become confluent (the adhesive surface of the plate is 100% of the cells), the number of cells is measured and subcultured to obtain a total of 16 types of cells. At the same time as measuring the growth rate of the cells, the morphology of the cells was observed under a microscope.
- the cell proliferation rate was measured over 80 days from the introduction of the immortalizing gene (number of cells at the time of introduction: 3 ⁇ 10 4 cells).
- the results (No. 1 to No. 16) arranged in descending order of the number of cells at 80 days are shown in Table 1 below together with the number of cells.
- the cell condition was observed at about 2 months (59th day) when the cell proliferation of the negative control stopped, and the uniformity of cell size and the degree of dead cells detached from the culture plate were observed in two stages. Judgment was made (Fig. 5). The judgment results are shown in Table 1 (cell size is uniform and cell peeling is small: ⁇ , cell size is non-uniform and cell peeling is large: ⁇ , presence or absence of immortalizing factor is present. Notated by + or-).
- FIG. 6 shows the cell growth curve.
- the growth stopped after about 2 months (66 days), but the growth of the immortalizing factor-introduced cells (No. 1, 2, 4, 12) was prolonged.
- the growth of hTERT gene-introduced cells (No. 12) was prolonged, the growth rate gradually decreased after 2 months, and further growth could not be expected.
- the combination-introduced cells (No. 1, 2, 4) of Bmi-1 and hTERT, hTERT and SV40T, and Bmi-1 and hTERT and SV40T proliferated stably without slowing down even on the 80th day. was observed.
- the growth rate of the combination of the three factors Bmi-1, hTERT and SV40T was the fastest.
- human umbilical cord matrix-derived mesenchymal stem cells product name: HumanMesenchymal Stem Cells from Umbilical Cord Matrix (hMSC-UC), Promo Cell, product code C-12971
- hMSC-UC HumanMesenchymal Stem Cells from Umbilical Cord Matrix
- Promo Cell product code C-12971
- Example 3 Analysis of immortalized cultured cells using a SeV vector carrying a 3-factor (Bmi-1 / hTERT / SV40T) gene (1) Preparation of 3-factor (Bmi-1 / hTERT / SV40T) -introduced immortalized cells In the examination of MSCs derived from different tissues, it was judged that the SeV vector carrying the three-factor (Bmi-1 / hTERT / SV40T) gene was the most suitable for immortalization, and detailed property analysis of the MSCs immortalized with this vector was performed. ..
- the cells used were MSCs derived from cord blood (product name: Umbilical Cord-Derived Mesenchymal Stem Cells; Normal, Human (ATCC PCS-500-010)) used in the selection of immortalizing factors, but the cells were longer than those used in the selection. Cultured (less than 1 month) cells were used. A 3-factor (Bmi-1 / hTERT / SV40T) gene-loaded SeV vector was introduced under the same MOI: 20 conditions as in Example 2. MSCs without gene transfer with SeV vector were simultaneously cultured as a negative control.
- the immortalized cell line introduced with 3 factors (Bmi-1 / hTERT / SV40T) was cultured in a CO 2 incubator at 35 ° C using a 6 cm dish, and the plates adhered. When the surface was grown to about 80% of the cells, 1/5 was subcultured to a new 6 cm dish, and the culture was continued for 75 days.
- the SeV vector carrying the immortalization gene is a temperature-sensitive vector, and it is possible to rapidly eliminate the intracellular SeV vector genome by raising the culture temperature from 35 ° C to 37 ° C (Efficient generation of transgene-free).
- human induced pluripotent stem cells (iPSCs) by temperature-sensitive Sendai virus vectors. Ban H, Nishishita N, Fusaki N, Tabata T, Saeki K, Shikamura M, Takada N, Inoue M, Hasegawa Natl Acad Sci U S A. 2011 Aug 23; 108 (34): 14234-9).
- Chromosome analysis of cells was performed by the quinacrine-Hoechst fractionation method at the point ⁇ in FIG.
- the chromosome slide is first immersed in 50 ml of Macylben solution (280 ml of 0.1 M citrate solution and 220 ml of 0.2 M disodium hydrogen phosphate solution are mixed and autoclaved), and then 50 ml of Macylben. It was soaked in a solution of Hoechst 33258 (cat: B-2883-25MG, Sigma) at 10 ng / ml for 30 minutes.
- the immortalized cell line was the parent strain at all points. It was a normal karyotype as well (Fig. 10).
- telomere length was evaluated at the points marked with * in FIGS. 6 and 7.
- the telomere length was evaluated by performing real-time PCR using genomic DNA as a template and by relative quantification of telomere sequences. Genomic DNA extraction from cells was performed using Gentra® Puregene® Kit (Qiagen) with reference to the manufacturer's procedure.
- Gentra® Puregene® Kit Qiagen
- the Telomere primer set and Single copy reference primer set attached to the Relative Human Telomere Length Quantification qPCR Assay Kit (ScienCell Research Laboratories) were used as primers, and FastStart Essential DNA Green Master (Roche) was used as the PCR reagent.
- the PCR reaction conditions were as recommended by the manufacturer. StepOnePlus (Life Technologies Japan) was used for PCR reaction and data acquisition, and data analysis was performed with StepOne Software v2.3.
- Example 4 Differentiation ability of immortalized population cells Regarding the immortalized cells (MSC) introduced with the three factors (Bmi-1 / hTERT / SV40T) prepared in Example 3, fat cells, osteoblasts, nerve cells, and chondrocytes. The ability to differentiate into cells was investigated.
- MSC (ATCC PCS-500-010) derived from cord blood that was immortalized with three factors of Bmi-1, hTERT, and SV40T and cultured for a long period of time (cultured for 4 months after gene transfer) was used. Four months is a difficult period for culturing in general MSCs that are not immortalized.
- a commercially available (PromoCell) differentiation kit was used for MSC differentiation.
- Mesenchymal Stem Cell Adipogenic Differentiation Medium 2 (product code C-28016) for adipose cell differentiation
- Mesenchymal Stem Cell Osteogenic Differentiation Medium (product code C-28013) for osteoblast differentiation
- Mesenchymal Stem Cell Neurogenic Differentiation for nerve cell differentiation.
- Medium (product code C-28015) and Mesenchymal Stem Cell Chondrogenic Differentiation Medium (product code C-28012) were used for cartilage cell differentiation, and the operation method was carried out according to the attached protocol.
- the confirmation of adipocyte differentiation was performed using the fluorescent dye Lipi-Green (Doujin Kagaku: product code LD02), which specifically stains lipid droplets, which is an index of adipocyte differentiation. Osteoblast differentiation was confirmed by alkaline phosphatase staining. Nerve cell differentiation was confirmed using the fluorescent dye NeuroFluor NeuO (VERITAS: product code ST-01801), which specifically stains nerve cells. Chondrocyte differentiation was confirmed by alcian blue staining.
- Lipi-Green Doujin Kagaku: product code LD02
- Osteoblast differentiation was confirmed by alkaline phosphatase staining.
- Nerve cell differentiation was confirmed using the fluorescent dye NeuroFluor NeuO (VERITAS: product code ST-01801), which specifically stains nerve cells. Chondrocyte differentiation was confirmed by alcian blue staining.
- MSCs that retain the differentiation potential and have not been gene-introduced in the logarithmic growth phase were used as positive controls, and compared with the differentiation potential of immortalized MSCs that had passed 4 months, fat.
- lipid droplets which are indicators of differentiation, were observed in both cases.
- the immortalized MSC had a lower degree of differentiation than the positive control (Fig. 12).
- osteoblast differentiation staining with alkaline phosphatase was confirmed in both immortalized MSC and positive control, and the cells were differentiated into osteoblasts (Fig. 13).
- Immortalized MSCs had a higher differentiation rate than positive controls.
- Example 5 Cloning test of immortalized MSCs For the three-factor (Bmi-1 / TERT / SV40T) -introduced immortalized cells (MSCs) prepared in Example 3, whether cloning is possible or whether the MSCs are not immortalized We conducted a confirmation experiment to see how difficult cloning is.
- MSC (ATCCPCS-500-010) derived from umbilical cord blood that retains differentiation potential and has not been gene-introduced in the logarithmic growth phase in early cells cultured for about 2 weeks after purchase, has just been gene-introduced ( Cloning was performed using three types of MSCs: an immortalized MSC (2 weeks old) and an immortalized MSC (4 months old) cultured for a long time after gene transfer.
- the three types of MSCs that had been cultured and maintained were once completely separated into single cells, and the number of cells was measured.
- the cells were seeded at a variable number such as 32 cells / 100 ⁇ l, and cultured in a 5% CO 2 incubator at 35 ° C until single cells formed colonies. For medium exchange, half the amount was exchanged every 3 days so as not to disturb colonization.
- Non-immortalized early growth MSCs corresponded to common MSCs, but only 2 wells of 32 cells / well compartments were formed.
- MSCs into which the immortalizing gene was introduced with the SeV vector colonies were formed at a high frequency of 9 well / 24 well even at a low density of 4 cells / well in the two weeks immediately after the introduction (Fig. 16). After 4 months, colonies were formed in 16 wells / 24 wells and more than half of the wells even at a low density of 4 cells / well (Fig. 17).
- Example 6 Various analyzes of clone immortalized MSC (1) Proliferative ability of cloned cells Five clones of MSC cloned as a single cell 2 weeks after infection with SeV vector were divided into 35 ° C and 37 ° C after culturing for 60 days. Was cultured. Three of the five clones proliferated and decreased with the removal of the SeV vector at 37 ° C. A growth curve is shown for clone A10 among them (Fig. 18).
- Example 7 Immortalization and single-cell cloning of MSCs with different cell ages (aging degree) Since the cell populations grown from one MSC have the same genetic properties, the quality is constant and quality control becomes easy. In that respect, single-cell cloning has important significance in regenerative medicine, and single-cell cloning is indispensable in that when a gene is introduced from the outside, only the target cell is selected.
- MSC isolated from the human body is a mixture of cells of various stages, from young cells to aged cells. The ratio also varies from individual to individual, and it is difficult to extract and proliferate only cells at a specific time (youth). Conversely, if MSCs can be immortalized at any time from young cells to senescent cells and can be further cloned, their application range will be greatly expanded in regenerative medicine.
- the cells used were MSCs derived from umbilical cord blood (product name: Umbilical Cord-Derived Mesenchymal Stem Cells; Normal, Human (ATCC PCS-500-010)).
- the purchased cells were cultured for a long period of about 80 days at 37 ° C until proliferation stopped. During that time, a part of the cells was cryopreserved at about every other week of passage (Fig. 21).
- the cells were transferred to a CO 2 incubator at 37 ° C to 35 ° C and expanded and cultured for 8 days (24 well ⁇ 6 well). After that, the immortalized cells were seeded on a 96-well plate so that the number of cells per well was 1, 2, or 4, and the number of non-immortalized control cells was 5, 10 per well. The cells were seeded to 15 or 20 cells, cultured for about 2 weeks until one cell divided and proliferated and a cell population (colony) appeared, and the number of colonies generated by microscopic observation was counted.
- Example 8 Confirmation of effect on MSCs in which proliferation has stopped The effect of the SeV vector carrying an immortalizing gene on MSCs in which cell division has completely stopped due to aging was confirmed.
- SeV vector non-infected cells control cells
- SeV vector-infected cells no change was observed for several days, but after one week, many cells changed roundly and seemed to die as they were, but small adhesive cells were observed. After 2 weeks, the cells changed to a vigorously proliferating cell population similar to immortalized cells (Fig. 24).
- Infected cells and infected cells were cultured in a medium containing serum from the day of infection to the 11th and 17th days, respectively.
- a medium having a composition of D-MEM (Low Gulucose), 20% FBS, 0.01 mol / L Hepes, Penicillin 100units / ml, Streptomycin 100 ⁇ g / ml, and bFGF 20 ng / ml was used. Then, it was replaced with a serum-free medium and the culture was continued.
- Stem Fit For Mesenchymal Stem Cell (AJINOMOTO; A3) medium was used as the serum-free medium.
- As the culture dish a 6 cm dish (CORNING; 353004) coated with iMatrix-511 silk (Matrixome; 892 091) was used.
- Infection with the SeV vector was performed on 2 ⁇ 10 5 hMSC-AT cells or hMSC-BM cells under the condition of MOI: 40.
- the non-infection condition and the infection condition were carried out once each.
- the medium was changed 24 hours after infection and maintenance culture was continued.
- Cell culture was performed in a 5% CO 2 incubator at 35 ° C.
- the non-infected cell line was found to have a decrease in the number of cells, so the culture was terminated 52 days after the day of infection.
- the SeV vector-infected cell line continued to proliferate after the 52nd day (Fig. 26).
- Example 10 Examination of cell proliferation of immortalized rat MSC Rat subcutaneous fat-derived mesenchymal stem cells rMSC cells (Cosmo Bio Co., Ltd., MSA01C) are media for proliferation of rat subcutaneous fat-derived mesenchymal stem cells (Cosmo Bio Co., Ltd.). Cultivated in MSA-GM).
- Infection with the SeV vector was performed on 2 ⁇ 10 5 rMSC cells under the condition of MOI: 40.
- the non-infected condition was once and the infected cell line was twice.
- the medium was changed 24 hours after infection and maintenance culture was continued.
- Culture dishes were placed in a collagen coated dish (IWAKI, 4810-010) and maintained at 35 ° C. in a 5% CO 2 incubator.
- cells with advanced cellular senescence may exhibit characteristics such as an increase in cell size, a flat shape, and the formation of vacuoles.
- Infected cells cultured at 35 ° C showed no characteristic of cellular senescence compared to infected cells cultured at 37 ° C. From this result, it was confirmed that the cells continued to be cultured at 35 ° C. did not undergo cell senescence in terms of cell morphology.
- Example 12 Examination of cell proliferation of immortalized HUVEC Human umbilical vein endothelial cells (HUVEC cells, Promocell, C-12205) were cultured in an endothelial cell medium (ScienCell; 1001).
- Infection with the SeV vector was performed on 2 ⁇ 10 5 HUVEC cells under MOI: 40 conditions. The non-infection condition was once and the infection condition was twice. The medium was changed 24 hours after infection and maintenance culture was continued. Infected cells were divided into two conditions, one was to culture at 35 ° C and the other was to culture at 37 ° C on the 35th day after infection (Fig. 33, arrow). Cell culture was performed at each temperature in a 5% CO 2 incubator.
- the non-infected cells were found to have a decrease in the number of cells, so the culture was terminated on the 39th day from the day of infection. On the other hand, the infected cells continued to proliferate even 74 days after the infection (Fig. 33). From this, it was confirmed that cell proliferation can be prolonged even in HUVEC cells by SeV vector infection.
- Example 14 Verification of enteritis healing effect of immortalized MSC in an inflammatory bowel disease model (1) CD4 positive CD45RB high positive CD25 negative (CD4 + CD45RB High + CD25- ) Isolation of T cells 8 weeks old BALB / cAJcl ( After acclimatization of 30 females of Claire Japan) for 1 week, blood was removed by cardiac blood sampling under anesthesia, and the spleen and mesenteric lymph nodes were collected. The spleen was hemolyzed after tissue dispersion using the MACS system (Miltenyi Biotech). The mesenteric lymph nodes were crushed with a 1 mL syringe plunger and filtered through a 40 ⁇ m cell strainer.
- Antibodies to isolate CD4 + CD45RB High + CD25-T cells after combining spleen and mesenteric lymph node cells and separating CD4 positive cells by CD4 microbeads (Miltenyi Biotech) treatment. Labeled with mouse CD4 antibody (BD), FITC Rat Anti-Mouse CD45RB antibody (BD), PE / Cy7 anti-mouse CD25 antibody (BioLegend)) and fractionated by flow cytometer (MoFlo XDP, BECKMAN).
- CD4 + CD45RB High + CD25 --T cell transfer 9-week-old female SCID CB-17 / lcr-scid / scidJcl: Japan Marie
- Cells were transplanted by tail vein injection (cell transplant group: 1 group 8 animals x 5 groups, untreated group (PBS administration): 1 group x 7 animals). After transplantation, body weight was measured three times a week and the coat and stool condition were observed. Twenty-one days after cell administration, randomization was performed using the "multivariable block allocation" system of the statistical analysis software JMP based on body weight changes, and the cell transplantation group was divided into groups. At that time, individuals who deviated from the average relative body weight of ⁇ 2 SD were excluded.
- hMSC-UC parent MSC
- immortalized MSC were cultured under each cell culture condition.
- MSC administration (therapeutic cell transplantation)
- parental MSC and immortalized MSC were administered 1.0 ⁇ 10 6 per mouse by tail vein injection.
- Dexamethasone (Sigma): Dex (1 mg / kg, 100 ⁇ L / animal) was subcutaneously administered from the 21st day to the 14th day. Dex solution was prepared at the time of use based on the group average body weight on the 21st and 28th days.
- the present invention can be used in the fields of cell medicine and regenerative medicine.
- the cell immortalization technique of the present invention can be used not only for normal cells with slow cell proliferation but also for cancer cells and the like, and can also be used in the field of basic research. Furthermore, single-cell cloning, which is essential for gene transfer and chromosome transfer, is also possible.
- the cells obtained by the present invention continue to proliferate without aging, which facilitates quality control, promotes mechanization of culture, significantly reduces cell production costs, and handles a large number of cells. Is possible. Therefore, the present invention expands the range of applicable diseases in cell medicine and regenerative medicine, and contributes to the activation of domestic and foreign medical-related industries. All publications, patents and patent applications cited herein are incorporated herein by reference in their entirety.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Engineering & Computer Science (AREA)
- Genetics & Genomics (AREA)
- Biomedical Technology (AREA)
- Organic Chemistry (AREA)
- Chemical & Material Sciences (AREA)
- Zoology (AREA)
- Biotechnology (AREA)
- Wood Science & Technology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- General Engineering & Computer Science (AREA)
- Microbiology (AREA)
- Biochemistry (AREA)
- General Health & Medical Sciences (AREA)
- Developmental Biology & Embryology (AREA)
- Cell Biology (AREA)
- Hematology (AREA)
- Rheumatology (AREA)
- Virology (AREA)
- Physics & Mathematics (AREA)
- Biophysics (AREA)
- Molecular Biology (AREA)
- Plant Pathology (AREA)
- Immunology (AREA)
- Micro-Organisms Or Cultivation Processes Thereof (AREA)
Abstract
Description
[1]以下の工程を含む、可逆的不死化細胞の製造方法。
(1)不死化遺伝子を搭載する染色体非組み込み型RNAウイルスベクターを哺乳類の細胞に導入し、該細胞で不死化遺伝子を発現させる工程、及び
(2)工程(1)で得られた細胞を培養し、増殖させる工程
[2]前記不死化遺伝子が、Bmi-1遺伝子、TERT遺伝子、及びSV40T遺伝子よりなる群から選択される1種又は2種以上の不死化遺伝子である、[1]に記載の方法。
[3]前記不死化遺伝子が以下の(a)~(d)のいずれかである、[1]又は[2]に記載の方法。
(a) Bmi-1遺伝子、TERT遺伝子、及びSV40T遺伝子の組み合わせ
(b) Bmi-1遺伝子とTERT遺伝子の組み合わせ
(c) TERT遺伝子とSV40T遺伝子の組み合わせ
(d) TERT遺伝子
[4]前記細胞が、体細胞である、[1]~[3]のいずれかに記載の方法。
[5]前記体細胞が、体性幹細胞である、[4]に記載の方法。
[6]前記体性幹細胞が、間葉系幹細胞である、[5]に記載の方法。
[7]前記染色体非組み込み型RNAウイルスベクターが、マイナス鎖RNAウイルスベクターである、[1]~[6]のいずれかに記載の方法。
[8]前記マイナス鎖RNAウイルスベクターが、パラミクソウイルスベクターである、[7]に記載の方法。
[9]前記パラミクソウイルスベクターが、センダイウイルスベクターである、[8]に記載の方法。
[10]前記センダイウイルスベクターが、温度感受性センダイウイルスベクターである、[9]に記載の方法。
[11]前記染色体非組み込み型RNAウイルスベクターがセンダイウイルスベクターであり、前記工程(2)の培養後、該センダイウイルスベクターを除去する工程をさらに含む、[1]に記載の方法。
[12]前記センダイウイルスベクターの除去が、培養温度を35℃から37℃に変更することにより行われる、[11]に記載の方法。
[13]前記工程(2)の培養後、不死化細胞をクローニングする工程をさらに含む、[1]~[12]のいずれかに記載の方法。
[14][1]~[13]のいずれかに記載の方法により得られる不死化細胞。
[15]Bmi-1遺伝子、TERT遺伝子、及びSV40T遺伝子よりなる群から選択される1種又は2種以上の不死化遺伝子を搭載するセンダイウイルスベクターを除去可能な状態で含む、不死化細胞。
[16][14]又は[15]に記載の不死化細胞を含む再生医療用製品。
[17]Bmi-1遺伝子、TERT遺伝子、及びSV40T遺伝子よりなる群から選択される1種又は2種以上の不死化遺伝子を搭載する、温度感受性センダイウイルスベクター。
[18][17]に記載の温度感受性センダイウイルスベクターを含む、可逆的不死化細胞作製用キット。
本願は、2020年11月6日に出願された日本国特許出願2020-186146号の優先権を主張するものであり、該特許出願の明細書に記載される内容を包含する。 That is, the present invention includes the following inventions.
[1] A method for producing a reversible immortalized cell, which comprises the following steps.
(1) A step of introducing a chromosome-non-integrating RNA viral vector carrying an immortalizing gene into mammalian cells and expressing the immortalizing gene in the cells, and (2) culturing the cells obtained in step (1). And the process of growing
[2] The method according to [1], wherein the immortalizing gene is one or more immortalizing genes selected from the group consisting of the Bmi-1 gene, the TERT gene, and the SV40T gene.
[3] The method according to [1] or [2], wherein the immortalizing gene is any of the following (a) to (d).
(a) Combination of Bmi-1 gene, TERT gene, and SV40T gene
(b) Combination of Bmi-1 gene and TERT gene
(c) Combination of TERT gene and SV40T gene
(d) TERT gene
[4] The method according to any one of [1] to [3], wherein the cell is a somatic cell.
[5] The method according to [4], wherein the somatic cell is a somatic stem cell.
[6] The method according to [5], wherein the somatic stem cell is a mesenchymal stem cell.
[7] The method according to any one of [1] to [6], wherein the chromosomal non-integrated RNA viral vector is a negative strand RNA viral vector.
[8] The method according to [7], wherein the negative strand RNA virus vector is a paramyxovirus vector.
[9] The method according to [8], wherein the paramyxovirus vector is a Sendai virus vector.
[10] The method according to [9], wherein the Sendai virus vector is a temperature sensitive Sendai virus vector.
[11] The method according to [1], wherein the chromosomal non-integrating RNA virus vector is a Sendai virus vector, further comprising a step of removing the Sendai virus vector after culturing in the step (2).
[12] The method according to [11], wherein the removal of the Sendai virus vector is performed by changing the culture temperature from 35 ° C to 37 ° C.
[13] The method according to any one of [1] to [12], further comprising a step of cloning immortalized cells after culturing in the step (2).
[14] Immortalized cells obtained by the method according to any one of [1] to [13].
[15] Immortalized cells comprising a Sendai virus vector carrying one or more immortalizing genes selected from the group consisting of Bmi-1 gene, TERT gene, and SV40T gene in a removable state.
[16] A regenerative medicine product comprising the immortalized cell according to [14] or [15].
[17] A temperature-sensitive Sendai virus vector carrying one or more immortalizing genes selected from the group consisting of the Bmi-1 gene, the TERT gene, and the SV40T gene.
[18] A kit for producing reversible immortalized cells, which comprises the temperature-sensitive Sendai virus vector according to [17].
This application claims the priority of Japanese Patent Application No. 2020-186146 filed on November 6, 2020, and includes the contents described in the specification of the patent application.
本発明は、可逆的不死化細胞の製造方法であって、該方法は、(1)不死化遺伝子を搭載する染色体非組み込み型RNAウイルスベクターを哺乳類の細胞に導入し、該細胞で不死化遺伝子を発現させる工程、及び(2)工程(1)で得られた細胞を培養し、増殖させる工程を含む。 1. 1. Method for Producing Reversible Immortalized Cells The present invention is a method for producing reversible immortalized cells, wherein the method (1) introduces a chromosomal non-integrating RNA virus vector carrying an immortalizing gene into mammalian cells. Then, the step of expressing the immortalizing gene in the cell and the step of (2) culturing and proliferating the cell obtained in the step (1) are included.
本発明において、「細胞」とは、生殖系列細胞(卵子及び精子、卵母細胞、ES細胞など)及び分化全能性細胞(iPS細胞)以外の全ての体細胞をいう。また、「体細胞」は、初代培養細胞、継代細胞、及び株化細胞のいずれであってもよい。さらに、体細胞は、天然由来であっても、iPS細胞等より分化させて人工的に作製されたものであってもよい。体細胞としては、具体的には、組織を形成する細胞(脂肪細胞、線維芽細胞、神経細胞、皮膚細胞、血液細胞、筋肉細胞、骨芽細胞、軟骨細胞、肝細胞、膵細胞、腎細胞、心筋細胞、脳細胞、肺細胞、脾細胞、副腎細胞、歯肉細胞、歯根膜細胞)などの分化した細胞、又はそれらの前駆細胞等、免疫系の細胞(B細胞、T細胞、単球系の細胞等)、体性幹細胞[間葉系幹細胞(脂肪由来幹細胞、骨髄由来幹細胞、臍帯血由来幹細胞、胎盤由来幹細胞等)、造血幹細胞、神経幹細胞、表皮幹細胞、腸管上皮幹細胞、歯髄幹細胞、歯根膜幹細胞等]などが挙げられる。細胞の由来は哺乳動物であれば特に限定はされず、例えば、ヒト、マウス、ラット、モルモット、ハムスター、ウサギ、イヌ、ネコ、ブタ、ウシ、ウマ等が挙げられる。 (cell)
In the present invention, the "cell" refers to all somatic cells other than germline cells (egg and sperm, oocyte, ES cell, etc.) and totipotent cells (iPS cells). Further, the "somatic cell" may be any of a primary cultured cell, a subcultured cell, and an established cell line. Further, the somatic cells may be naturally derived or artificially produced by differentiating from iPS cells or the like. Specific examples of somatic cells include cells that form tissues (fat cells, fibroblasts, nerve cells, skin cells, blood cells, muscle cells, osteoblasts, cartilage cells, hepatocytes, pancreatic cells, and renal cells. , Myocardial cells, brain cells, lung cells, splenocytes, adrenal cells, gingival cells, root membrane cells) or their precursor cells, immune system cells (B cells, T cells, monocytic cells) (Cells, etc.), somatic stem cells [mesophageal stem cells (adipose-derived stem cells, bone marrow-derived stem cells, umbilical cord blood-derived stem cells, placenta-derived stem cells, etc.), hematopoietic stem cells, nerve stem cells, epidermal stem cells, intestinal epithelial stem cells, dental pulp stem cells, tooth roots, etc. Membrane stem cells, etc.] and the like. The origin of the cells is not particularly limited as long as it is a mammal, and examples thereof include humans, mice, rats, guinea pigs, hamsters, rabbits, dogs, cats, pigs, cows, and horses.
「不死化細胞」とは、初代培養細胞や通常の培養条件で培養された細胞とは異なり、細胞分裂を繰り返しても増殖が停止しない細胞、すなわち、無限増殖能を有する細胞をいう。本発明における「不死化細胞」とは、所定の不死化遺伝子の導入によって無限増殖が可能となった細胞で、かつ繰り返し継代して培養しても、その無限増殖能が低下しない細胞をいう。本発明に係る「不死化細胞」は、細胞の由来あるいは培養条件により増殖速度や増殖期間は異なるが、継代培養の結果、同一の培養条件下において、無処理の細胞が増殖低下又は停止する時期以降も、20日以上、好ましくは60日以上、より好ましくは80日以上指数増殖を続けることが可能である。また、本発明の不死化細胞は、上記のような無限増殖が可能な細胞集団、及び当該細胞集団よりクローニングされた不死化細胞株のいずれをも包含する。また、「可逆的不死化」とは、不死化遺伝子を細胞に導入して、当該細胞を無限に増殖可能な状態にした後、当該不死化遺伝子を除去することによって、細胞増殖を停止又は減弱させることをいう。 (Reversible immortalized cells)
The "immortalized cell" refers to a cell whose proliferation does not stop even if cell division is repeated, that is, a cell having an infinite proliferation ability, unlike a primary cultured cell or a cell cultured under normal culture conditions. The "immortalized cell" in the present invention means a cell capable of infinite proliferation by introducing a predetermined immortalizing gene, and a cell whose infinite proliferation ability does not decrease even if it is repeatedly subcultured and cultured. .. The "immortalized cells" according to the present invention have different growth rates and growth periods depending on the origin of the cells or the culture conditions, but as a result of subculture, untreated cells proliferate or cease under the same culture conditions. It is possible to continue exponential growth for 20 days or more, preferably 60 days or more, more preferably 80 days or more even after the period. In addition, the immortalized cells of the present invention include both the cell population capable of infinite proliferation as described above and the immortalized cell line cloned from the cell population. In addition, "reversible immortalization" means that a cell proliferation is stopped or attenuated by introducing an immortalizing gene into a cell, making the cell proliferative indefinitely, and then removing the immortalizing gene. It means to let.
本発明の不死化細胞は、細胞に所定の不死化遺伝子を染色体非組み込み型RNAウイルスベクターを用いて導入することによって作製される。ここで「不死化遺伝子」とは、細胞を不死化して無限増殖能を獲得させ、かつ細胞死を誘導しない遺伝子をいう。また、不死化遺伝子は、外因性遺伝子であり、細胞外から新たに導入される不死化遺伝子を意味する。さらに、不死化遺伝子は、ヒト以外に由来する不死化遺伝子であってもよく、標的細胞内で発現可能な形態に改変された不死化遺伝子であってもよい。本発明においては、不死化遺伝子として、Bmi-1遺伝子、TERT遺伝子、及びSV40T遺伝子よりなる群から選択される1種又は2種以上の遺伝子を用いることができるが、2種の遺伝子の組み合わせが好ましく、3種の遺伝子の組み合わせがより好ましい。1種の遺伝子を用いる場合はTERT遺伝子が好ましく、2種以上の遺伝子を組み合わせる場合の好ましい組み合わせの例としては、Bmi-1遺伝子とTERT遺伝子の組み合わせ、TERT遺伝子とSV40T遺伝子の組み合わせが挙げられる。 (Immortalization gene)
The immortalized cell of the present invention is produced by introducing a predetermined immortalizing gene into a cell using a chromosome non-integrating RNA viral vector. Here, the "immortalizing gene" refers to a gene that immortalizes a cell to acquire infinite proliferation ability and does not induce cell death. The immortalizing gene is an extrinsic gene and means an immortalizing gene newly introduced from the outside of the cell. Further, the immortalizing gene may be an immortalizing gene derived from other than human, or may be an immortalizing gene modified into a form expressible in a target cell. In the present invention, as the immortalizing gene, one or more genes selected from the group consisting of the Bmi-1 gene, the TERT gene, and the SV40T gene can be used, but a combination of the two genes can be used. Preferably, a combination of three genes is more preferred. When one kind of gene is used, the TERT gene is preferable, and examples of a preferable combination when two or more kinds of genes are combined include a combination of Bmi-1 gene and TERT gene, and a combination of TERT gene and SV40T gene.
本発明においては、上記不死化遺伝子を細胞に導入し、発現させるためのベクターとして「染色体非組み込み型RNAウイルスベクター」を用いる。本発明において、ウイルスベクターとは、当該ウイルスに由来するゲノム核酸を有し、該核酸に導入遺伝子を組み込むことにより、該遺伝子を発現させることができるベクターを意味する。また、「染色体非組み込み型RNAウイルスベクター」は、ウイルスに由来し、遺伝子を標的細胞に導入することができるウイルスベクターであって、導入された遺伝子が宿主の染色体(核由来染色体)に組み込まれる危険性のない運搬体のことをいう。 (Chromosome non-integrated RNA viral vector)
In the present invention, a "chromosome non-integrating RNA virus vector" is used as a vector for introducing and expressing the immortalization gene in cells. In the present invention, the viral vector means a vector having a genomic nucleic acid derived from the virus and capable of expressing the gene by incorporating a transgene into the nucleic acid. The "chromosome non-integrated RNA virus vector" is a virus vector derived from a virus and capable of introducing a gene into a target cell, and the introduced gene is integrated into a host chromosome (nuclear-derived chromosome). A non-hazardous carrier.
本発明の好ましい態様において、染色体非組み込み型RNAベクターとして、センダイウイルス(Sendai virus, SeV)ベクターを用いる。前記Bmi-1遺伝子、TERT遺伝子、及びSV40T遺伝子の各不死化因子遺伝子は別々にSeVベクターに挿入してもよく、また単一のSeVベクターに一緒に挿入してもよい。 (Sendai virus vector: SeV vector)
In a preferred embodiment of the present invention, a Sendai virus (SeV) vector is used as the non-chromosomal integrated RNA vector. The immortalizing factor genes of the Bmi-1 gene, TERT gene, and SV40T gene may be inserted separately into the SeV vector, or may be inserted together into a single SeV vector.
不死化遺伝子(Bmi-1遺伝子、TERT遺伝子、及びSV40T遺伝子)が組み込まれる位置は特に制限されないが、各不死化遺伝子を別個の複数のベクターに挿入する場合、Bmi-1遺伝子は、NP遺伝子の上流、TERT遺伝子は、P遺伝子とM遺伝子の間、SV40T遺伝子は、NP遺伝子の上流に挿入することが好ましい。また2つ以上(Bmi-1とTERT、TERTとSV40T、又はBmi-1とTERTとSV40T)の遺伝子が単一のベクターに挿入されていてもよい。 (Construction of SeV vector carrying immortalizing gene)
The position where the immortalizing gene (Bmi-1 gene, TERT gene, and SV40T gene) is integrated is not particularly limited, but when each immortalizing gene is inserted into multiple separate vectors, the Bmi-1 gene is the NP gene. Upstream, the TERT gene is preferably inserted between the P and M genes, and the SV40T gene is preferably inserted upstream of the NP gene. Also, two or more genes (Bmi-1 and TERT, TERT and SV40T, or Bmi-1 and TERT and SV40T) may be inserted into a single vector.
上記のようにして得られる不死化遺伝子を搭載したSeVベクターは、該ベクター(センダイウイルス粒子)を体細胞の培地に添加して該細胞にウイルスを感染させることにより、該細胞内に導入される。ベクターの用量は細胞の種類や細胞密度、培地の量によって異なるため、感染効率が100%に近いMOIを、使用する細胞毎に予め調査して決定すればよい。 (Introduction of immortalization gene into cells)
The SeV vector carrying the immortalization gene obtained as described above is introduced into the cells by adding the vector (Sendai virus particles) to the medium of the somatic cells and infecting the cells with the virus. .. Since the dose of the vector varies depending on the cell type, cell density, and amount of medium, the MOI whose infection efficiency is close to 100% may be investigated and determined in advance for each cell to be used.
本発明において不死化遺伝子を導入した細胞を培養する方法は、通常の哺乳動物の体細胞の培養の方法及び条件に従って行うことができる。培養に用いる培地は特に限定されず、細胞の維持培養又は拡大培養のために一般的に使用され、ウイルス感染に適した培地を用いればよく、市販の培地、自製した培地のいずれであってもよい。例えば、細胞の生存及び増殖に必要な成分(無機塩、炭水化物、ホルモン、必須アミノ酸、非必須アミノ酸、ビタミン、脂肪酸)を含む基本培地、具体的には、Dulbecco's Modified Eagle's Medium (D-MEM)培地、Dulbecco's Modified Eagle's Medium:Nutient Mixture F-12(D-MEM/F-12) 培地、Glasgow MEM(G-MEM)培地、Basal Medium Eagle (BME)培地、Minimum Essential Medium(MEM)培地、Eagle's minimal essential medium(EMEM)培地、Iscove's Modified Dulbecco's Medium (IMDM)培地、RPMI 1640培地、Medium 199培地、αMEM培地、ハム培地、Fischer培地、及びこれらの混合培地などが挙げられる。また、培地には、必要に応じて、増殖因子(FGF、EGF等)、インターロイキン類、インスリン、トランスフェリン、ヘパリン、ヘパラン硫酸、コラーゲン、フィブロネクチン、プロゲステロン、セレナイト、B27-サプリメント、N2-サプリメント、抗生物質(ペニシリン、ストレプトマイシン等)等を含有してもよい。また、培地は、血清含有培地であっても無血清培地であってもよい。異種動物由来成分の混入防止の観点からは血清を含有しないか、培養する細胞と同種動物由来の血清を用いることが好ましい。また、アルブミン等の血清代替物を用いてもよい。 (Culture of immortalized gene-introduced cells)
In the present invention, the method for culturing cells into which an immortalizing gene has been introduced can be carried out according to the method and conditions for culturing normal mammalian somatic cells. The medium used for culturing is not particularly limited, and a medium generally used for maintenance culture or expansion culture of cells and suitable for virus infection may be used, and either a commercially available medium or a self-made medium may be used. good. For example, a basal medium containing components necessary for cell survival and proliferation (inorganic salts, carbohydrates, hormones, essential amino acids, non-essential amino acids, vitamins, fatty acids), specifically Dulbecco's Modified Eagle's Medium (D-MEM) medium. , Dulbecco's Modified Eagle's Medium: Natural Mixture F-12 (D-MEM / F-12) Medium, Glassgow MEM (G-MEM) Medium, Basal Medium Eagle (BME) Medium, Minimum Essential Medium (MEM) Medium, Eagle's minimal essential Examples include medium (EMEM) medium, Iscove's Modified Dulbecco's Medium (IMDM) medium, RPMI 1640 medium, Medium 199 medium, αMEM medium, ham medium, Fischer medium, and a mixed medium thereof. In addition, the medium may contain growth factors (FGF, EGF, etc.), interleukins, insulin, transferase, heparin, heparan sulfate, collagen, fibronectin, progesterone, selenite, B27-supplement, N2-supplement, antibiotics, if necessary. It may contain a substance (penicillin, streptomycin, etc.) and the like. Further, the medium may be a serum-containing medium or a serum-free medium. From the viewpoint of preventing contamination of heterologous animal-derived components, it is preferable to use serum that does not contain serum or that is derived from the same animal as the cells to be cultured. Alternatively, a serum substitute such as albumin may be used.
本発明によれば、上記のようにして作製した不死化細胞を分化誘導培地で培養することにより特定の組織細胞に分化誘導することができる。例えば、不死化細胞が間葉系幹細胞である場合、脂肪細胞、骨芽細胞、神経細胞、軟骨細胞等に分化させることできる。 2. 2. Induction of Differentiation According to the present invention, the immortalized cells prepared as described above can be induced to differentiate into specific tissue cells by culturing them in a differentiation-inducing medium. For example, when the immortalized cell is a mesenchymal stem cell, it can be differentiated into adipocyte, osteoblast, nerve cell, chondrocyte and the like.
以降の実施例において、ヒト間葉系幹細胞(MSC:mesenchymal stem cell)を培養する血清含有培地は、D-MEM(Low Gulucose), 20%FBS, 0.01mol/L Hepes, Penicillin100units/ml, Streptomycin100μg/ml, bFGF 20ng/mlの組成で作製した。bFGFは非常に半減期が短いので、安定性に優れたGibco Heat Stable Recombinant Human bFGF(Thermo Fisher Scientific)を使用した。MSCは上記培地にて、5%CO2インキュベーター内で培養を行った。なお、実験に使用したヒトのMSCは、骨髄由来、脂肪組織由来、臍帯血由来、及び臍帯マトリックス由来のいずれかである。 Hereinafter, the present invention will be described in more detail based on examples, but the present invention is not limited to these examples.
In the following examples, the serum-containing medium for culturing human mesenchymal stem cells (MSC) is D-MEM (Low Gulucose), 20% FBS, 0.01 mol / L Hepes, Penicillin 100units / ml,
(1)不死化遺伝子搭載SeVベクターの作製
不死化因子として汎用されているBmi-1(B lymphoma Mo-MLV insertion region 1 homolog)、hTERT(human telomerase reverse transcriptase)、SV40T(simian virus 40 large T antigen)、E6/E7(human papillomavirus 16 E6 protein and E7 protein)を選択し、染色体非組み込み型RNAウイルスベクターのSeVベクターに、Bmi-1遺伝子(配列番号1)、hTERT遺伝子(配列番号2)、SV40T遺伝子(配列番号3)、E6/E7遺伝子(配列番号4)をそれぞれ組み込んだ。SeVベクターへの不死化遺伝子搭載とベクターの製造は、株式会社IDファーマ(本社:つくば市)に依頼した。また4種類のベクターのうち3種類のベクターについては、蛍光色素遺伝子のORF(赤色)、GFP(緑色)、BFP(青色)も同時に搭載し、遺伝子発現(ベクターゲノム)の有無が顕微鏡で容易に確認出来るようにした。図1に各ベクターの構造を示す。SeVベクターへ搭載する不死化遺伝子の位置については、高発現のほうが結果的に長期発現し、SeVベクターにおいてはベクターゲノムの上流に搭載したほうが高発現になるため、Bmi-1,SV40Tはベクターゲノムの最も上流に搭載した。hTERTとE6/E7は高発現に起因すると思われるベクター生産効率低下のため搭載位置を下流に変更した。 (Example 1) Preparation of seV vector carrying an immortalizing gene and examination of infection conditions (1) Preparation of a SeV vector carrying an immortalizing gene Bmi-1 (B lymphoma Mo-
SeVベクターで遺伝子導入する際は、細胞タイプの違いにより感染効率が大きく変化する。また、過剰に感染させると細胞の種類によっては障害が生じることもある。よって、使用する細胞種で事前に感染MOI(Multiplicity of Infection:1細胞に対するベクター粒子の数)を決定するため、最初にMSCでの感染MOIを検討した。 (2) Examination of SeV vector infection conditions When transfecting with SeV vector, the infection efficiency changes greatly depending on the cell type. In addition, over-infection may cause damage depending on the cell type. Therefore, in order to determine the infected MOI (Multiplicity of Infection: number of vector particles per cell) in advance for the cell type to be used, the infected MOI in MSC was first examined.
4種の不死化因子(Bmi-1, hTERT, SV40T, E6/E7)遺伝子を単独搭載したSeVベクターをMSCに感染させ、長期間培養することにより、MSCの不死化に必要な因子を選定した。 (Example 2) Selection of immortalizing factor MSC by infecting MSC with a SeV vector carrying four kinds of immortalizing factor (Bmi-1, hTERT, SV40T, E6 / E7) genes alone and culturing for a long period of time. The factors necessary for immortalization of the disease were selected.
(1)3因子(Bmi-1/hTERT/SV40T)導入不死化細胞の作製
3種類の異なる組織由来のMSCの検討において、3因子(Bmi-1/hTERT/SV40T)遺伝子搭載SeVベクターが最も不死化に適していると判断し、このベクターで不死化したMSCの詳しい性状解析を行った。細胞は不死化因子の選定で用いた臍帯血由来のMSC(製品名:Umbilical Cord-Derived Mesenchymal Stem Cells; Normal, Human(ATCC PCS-500-010))を使用したが、選定の際よりも長期培養(1か月弱)した細胞を用いた。3因子(Bmi-1/hTERT/SV40T)遺伝子搭載SeVベクターで実施例2と同様にMOI:20の条件で遺伝子導入した。SeVベクターで遺伝子導入していないMSCをネガティブコントロールとして同時に培養した。 (Example 3) Analysis of immortalized cultured cells using a SeV vector carrying a 3-factor (Bmi-1 / hTERT / SV40T) gene (1) Preparation of 3-factor (Bmi-1 / hTERT / SV40T) -introduced immortalized cells In the examination of MSCs derived from different tissues, it was judged that the SeV vector carrying the three-factor (Bmi-1 / hTERT / SV40T) gene was the most suitable for immortalization, and detailed property analysis of the MSCs immortalized with this vector was performed. .. The cells used were MSCs derived from cord blood (product name: Umbilical Cord-Derived Mesenchymal Stem Cells; Normal, Human (ATCC PCS-500-010)) used in the selection of immortalizing factors, but the cells were longer than those used in the selection. Cultured (less than 1 month) cells were used. A 3-factor (Bmi-1 / hTERT / SV40T) gene-loaded SeV vector was introduced under the same MOI: 20 conditions as in Example 2. MSCs without gene transfer with SeV vector were simultaneously cultured as a negative control.
3因子(Bmi-1/hTERT/SV40T)導入不死化細胞株は、6cm dishを使用して35℃のCO2インキュベーター内で培養し、プレートの接着面が細胞で80%程度になるまで増殖した時期に新しい6cm dishに1/5を継代する方法で、75日間培養を続けた。 (2) Comparison of growth rate by change in culture temperature The immortalized cell line introduced with 3 factors (Bmi-1 / hTERT / SV40T) was cultured in a CO 2 incubator at 35 ° C using a 6 cm dish, and the plates adhered. When the surface was grown to about 80% of the cells, 1/5 was subcultured to a new 6 cm dish, and the culture was continued for 75 days.
温度変化から10日後に蛍光顕微鏡で蛍光タンパク質の発現を確認したところ、OFP(Bmi-1同時搭載)とGFP(hTERT同時搭載)の蛍光は、培養温度を35℃に維持した細胞においては確認されたものの、37℃に上げた細胞ではその発現がほとんど確認出来なくなった(図8)。 (3) Comparison of fluorescent protein expression and cell morphology due to changes in culture temperature When the expression of fluorescent protein was confirmed with a
細胞の染色体解析をキナクリン・ヘキスト分染法により、図7の▽のポイントで行った。キナクリン・ヘキスト分染法は、まず、50mlのマキルベン溶液(0.1M クエン酸溶液 280 mlと0.2M リン酸水素二ナトリウム溶液 220mlを混合し、オートクレーブする)に染色体スライドを浸し、ついで50 mlのマキルベン溶液に10ng/mlになるようにHoechst 33258(cat : B-2883-25MG, Sigma)を溶解したものに30分間浸した。染色体スライドの裏面から水流の弱い水道水を伝わらせて洗浄後、再びマキルベン溶液に5分間浸した後、マキルベン封入液(マキルベンとグリセロールを1:1で混合する)を用いてカバーガラスで覆い封入した。染色体解析顕微鏡(型式:AxioImager Z2, ZEISS)及び染色体解析ソフト(型式:Ikaros V5.7.4 CM/V5.4.12, Metasystems)を用いて解析を行った結果、不死化細胞株は、すべてのポイントで親株と同様に正常核型であった(図10)。 (4) Chromosome analysis Chromosome analysis of cells was performed by the quinacrine-Hoechst fractionation method at the point ▽ in FIG. In the quinacrine-hexist fractionation method, the chromosome slide is first immersed in 50 ml of Macylben solution (280 ml of 0.1 M citrate solution and 220 ml of 0.2 M disodium hydrogen phosphate solution are mixed and autoclaved), and then 50 ml of Macylben. It was soaked in a solution of Hoechst 33258 (cat: B-2883-25MG, Sigma) at 10 ng / ml for 30 minutes. After washing with tap water with a weak water flow from the back of the chromosome slide, soak it in the Makiruben solution again for 5 minutes, and then cover it with a Makiruben encapsulant (mixing Makiruben and glycerol 1: 1) with a cover glass. bottom. As a result of analysis using a chromosome analysis microscope (model: AxioImager Z2, ZEISS) and chromosome analysis software (type: Ikaros V5.7.4 CM / V5.4.12, Metasystems), the immortalized cell line was the parent strain at all points. It was a normal karyotype as well (Fig. 10).
テロメア長の評価を図6及び図7の*のポイントで行った。テロメア長の評価は、ゲノムDNAを鋳型にリアルタイムPCRを実施し、テロメア配列の相対定量により行った。細胞からのゲノムDNA抽出はGentra(登録商標)Puregene(登録商標)Kit (Qiagen)を用い、メーカーの手順を参照し実施した。リアルタイムPCRについて、プライマーはRelative Human Telomere Length Quantification qPCR Assay Kit (ScienCell Research Laboratories)付属のTelomere primer setとSingle copy reference primer setを用い、PCR試薬はFastStart Essential DNA Green Master (Roche)を用いた。PCR反応条件はメーカーの推奨条件に従った。PCR反応とデータ取得はStepOnePlus (ライフテクノロジーズジャパン)を使用し、StepOne Software v2.3にてデータ解析を行った。 (5) Evaluation of telomere length The telomere length was evaluated at the points marked with * in FIGS. 6 and 7. The telomere length was evaluated by performing real-time PCR using genomic DNA as a template and by relative quantification of telomere sequences. Genomic DNA extraction from cells was performed using Gentra® Puregene® Kit (Qiagen) with reference to the manufacturer's procedure. For real-time PCR, the Telomere primer set and Single copy reference primer set attached to the Relative Human Telomere Length Quantification qPCR Assay Kit (ScienCell Research Laboratories) were used as primers, and FastStart Essential DNA Green Master (Roche) was used as the PCR reagent. The PCR reaction conditions were as recommended by the manufacturer. StepOnePlus (Life Technologies Japan) was used for PCR reaction and data acquisition, and data analysis was performed with StepOne Software v2.3.
実施例3で作製した3因子(Bmi-1/hTERT/SV40T)導入不死化細胞(MSC)について、脂肪細胞、骨芽細胞、神経細胞、軟骨細胞へ分化への分化能を調べた。分化試験には、Bmi-1とhTERTとSV40Tの3因子で不死化し、長期にわたり培養した(遺伝子導入から4カ月培養した)臍帯血由来のMSC(ATCC PCS-500-010)を使用した。4カ月は不死化していない一般のMSCでは培養困難な期間である。 (Example 4) Differentiation ability of immortalized population cells Regarding the immortalized cells (MSC) introduced with the three factors (Bmi-1 / hTERT / SV40T) prepared in Example 3, fat cells, osteoblasts, nerve cells, and chondrocytes. The ability to differentiate into cells was investigated. For the differentiation test, MSC (ATCC PCS-500-010) derived from cord blood that was immortalized with three factors of Bmi-1, hTERT, and SV40T and cultured for a long period of time (cultured for 4 months after gene transfer) was used. Four months is a difficult period for culturing in general MSCs that are not immortalized.
実施例3で作製した3因子(Bmi-1/TERT/SV40T)導入不死化細胞(MSC)について、クローニングが可能かどうか、あるいは不死化していないMSCがどの程度クローニングが難しいのか確認実験を行った。 (Example 5) Cloning test of immortalized MSCs For the three-factor (Bmi-1 / TERT / SV40T) -introduced immortalized cells (MSCs) prepared in Example 3, whether cloning is possible or whether the MSCs are not immortalized We conducted a confirmation experiment to see how difficult cloning is.
(1)クローン細胞の増殖能
SeVベクター感染2週間後にシングルセルにしてクローニングした5クローンのMSCについて、60日間培養後に、35℃と37℃に分けて培養した。5クローンのうち3クローンは、37℃でSeVベクターの除去とともに増殖低下した。そのうちのクローンA10について増殖曲線を示す(図18)。 (Example 6) Various analyzes of clone immortalized MSC (1) Proliferative ability of cloned cells Five clones of MSC cloned as a
35℃においては、集団培養実験と同様にSeVベクターの存在は蛍光観察で確認できたが、37℃のグループはSeVベクターの存在は確認出来なかった。 (2) Confirmation of removal of SeV vector due to temperature change At 35 ° C, the presence of SeV vector could be confirmed by fluorescence observation as in the group culture experiment, but the presence of SeV vector could not be confirmed in the group at 37 ° C.
クローニングした10クローンについて染色体解析を行った結果、8クローンが正常核型であった。その中で図18に示した細胞増殖を示すクローンA10について、核型解析の結果を示す(図19)。 (3) Chromosome analysis of cloned cells As a result of chromosomal analysis of 10 cloned clones, 8 clones were normal karyotype. Among them, the results of karyotype analysis are shown for clone A10 showing cell proliferation shown in FIG. 18 (FIG. 19).
クローニングした不死化MSCのうち、5クローンについて実施例4と同じ方法で、脂肪細胞・骨芽細胞・神経細胞への分化能を調べた。その結果、クローンによって形態に違いが見られたのと同様に、分化程度にも違いが認められた。しかしいずれのクローンにおいても脂肪細胞・骨芽細胞・神経細胞への分化が認められ、多分化能が維持されていた(図20)。クローンによっては、クローニングする前の細胞より分化状態の非常に良いものもあり、神経分化においては、神経マーカーの発現だけでなく、神経突起が網目状に広がった神経ネットワークを形成している様子も観察された。 (4) Differentiation ability of cloned immortalized MSCs Among the cloned immortalized MSCs, 5 clones were examined for their ability to differentiate into fat cells, osteoblasts, and nerve cells by the same method as in Example 4. As a result, the degree of differentiation was also different, just as the morphology was different depending on the clone. However, in all the clones, differentiation into adipocytes, osteoblasts, and nerve cells was observed, and pluripotency was maintained (Fig. 20). Some clones are much better differentiated than the cells before cloning, and in neural differentiation, not only the expression of neural markers but also the appearance of neurites forming a network of neural networks. Observed.
1個のMSCから増殖した細胞集団は遺伝的性質が同じため、品質が一定になり、品質管理が容易になる点においてシングルセルクローニングは再生医療において重要な意味を持ち、また、外部から遺伝子導入した場合、目的細胞のみ選択する点においても、シングルセルクローニングは不可欠である。 (Example 7) Immortalization and single-cell cloning of MSCs with different cell ages (aging degree) Since the cell populations grown from one MSC have the same genetic properties, the quality is constant and quality control becomes easy. In that respect, single-cell cloning has important significance in regenerative medicine, and single-cell cloning is indispensable in that when a gene is introduced from the outside, only the target cell is selected.
老化し、完全に細胞分裂が停止したMSCに対する不死化遺伝子を搭載したSeVベクターの効果を確認した。 (Example 8) Confirmation of effect on MSCs in which proliferation has stopped The effect of the SeV vector carrying an immortalizing gene on MSCs in which cell division has completely stopped due to aging was confirmed.
脂肪組織由来ヒト間葉系幹細胞hMSC-AT(PromoCell; C-12977)及び骨髄由来ヒト間葉系幹細胞hMSC-BM(PromoCell; C-12974)を実験に使用した。hMSC-AT及びhMSC-BMのSeVベタクー感染実施日を「感染日」とすると、hMSC-AT非感染細胞と感染細胞は、それぞれ感染日より13日目と11日目まで、hMSC-BMの非感染細胞と感染細胞は、それぞれ感染日から11日目と17日目まで血清入り培地で培養した。血清入り培地は、D-MEM(Low Gulucose), 20%FBS, 0.01mol/L Hepes, Penicillin100units/ml, Streptomycin100μg/ml, bFGF 20ng/mlの組成の培地を使用した。その後、無血清培地に置換して培養を継続した。無血清培地には、Stem Fit For Mesenchymal Stem Cell(AJINOMOTO; A3)培地を使用した。培養皿は、iMatrix-511 silk(マトリクソーム; 892 091)でコーティングした6cm dish(CORNING; 353004)を使用した。 (Example 9) Examination of cell proliferation of human mesenchymal stem cells in serum-free medium Adipose tissue-derived human mesenchymal stem cells hMSC-AT (PromoCell; C-12977) and bone marrow-derived human mesenchymal stem cells hMSC-BM (PromoCell) C-12974) was used in the experiment. Assuming that the SeV betaku infection date of hMSC-AT and hMSC-BM is the "infection date", hMSC-AT non-infected cells and infected cells are non-infected cells of hMSC-BM from the infection date to the 13th and 11th days, respectively. Infected cells and infected cells were cultured in a medium containing serum from the day of infection to the 11th and 17th days, respectively. As the serum-containing medium, a medium having a composition of D-MEM (Low Gulucose), 20% FBS, 0.01 mol / L Hepes, Penicillin 100units / ml,
ラット皮下脂肪由来間葉系幹細胞rMSC細胞(コスモ・バイオ株式会社, MSA01C)はラット皮下脂肪由来間葉系幹細胞増殖用培地(コスモ・バイオ株式会社, MSA-GM)で培養した。 (Example 10) Examination of cell proliferation of immortalized rat MSC Rat subcutaneous fat-derived mesenchymal stem cells rMSC cells (Cosmo Bio Co., Ltd., MSA01C) are media for proliferation of rat subcutaneous fat-derived mesenchymal stem cells (Cosmo Bio Co., Ltd.). Cultivated in MSA-GM).
ヒト線維芽細胞(HFL1細胞,理研:RCB0521)はハム-F12培地(Nakalai; 17458-65)に、非働化したFetal bovine serum(NICHIREI; 175012)を15%(v/v)、ペニシリン-ストレプトマイシン溶液(富士フィルム和光; 168-23191)を100units/mLになるように添加した培地で培養した。 (Example 11) Examination of cell proliferation of immortalized HFL1 Human fibroblasts (HFL1 cells, RIKEN: RCB0521) were deactivated in ham-F12 medium (Nakalai; 17458-65) (NICHIREI; 175012). Was cultured in a medium containing 15% (v / v) of penicillin-streptomycin solution (Fuji Film Wako; 168-23191) at 100 units / mL.
ヒト臍帯静脈内皮細胞(HUVEC細胞,Promocell社,C-12205)は内皮細胞用培地(ScienCell; 1001)で培養した。 (Example 12) Examination of cell proliferation of immortalized HUVEC Human umbilical vein endothelial cells (HUVEC cells, Promocell, C-12205) were cultured in an endothelial cell medium (ScienCell; 1001).
染色体供与細胞としてWatanabe at al.(Mol Ther Nucleic Acids. 2015)に記載のHGF (hgf), GDNF (gdnf), IGF-1 (igf-1), 及びluciferase (e-luc)が搭載された21HAC2を保持するCHO細胞を用い、染色体受容細胞としては実施例1記載の不死化MSC細胞であるhMSC-UC No.3細胞を用い、Katoh et al. (BMC Biotechnology, 2010, 10:37)に記載の方法で微小核細胞融合及び培養を行った。BS選択培養下で1週間培養すると、耐性コロニーが出現し、4回の融合で得た合計9個のコロニーを単離し増殖させ、以後の解析を行った。 (1) Chromosome transfer by fusion of micronuclear cells and isolation of drug-resistant clones HGF (hgf), GDNF (gdnf), IGF-1 (Hgf), GDNF (gdnf), IGF-1 described in Watanabe at al. (Mol Ther Nucleic Acids. 2015) as chromosome-donating cells. CHO cells carrying 21HAC2 carrying igf-1), and luciferase (e-luc) were used, and hMSC-UC No. 3 cells, which are immortalized MSC cells described in Example 1, were used as chromosome receptor cells. , Katoh et al. (BMC Biotechnology, 2010, 10:37). Micronuclear cell fusion and culture were performed by the method described. After culturing under BS selective culture for 1 week, resistant colonies appeared, and a total of 9 colonies obtained by 4 fusions were isolated and proliferated, and the subsequent analysis was performed.
(2-1)蛍光顕微鏡観察
クローニングした9個のコロニーを蛍光顕微鏡下にて観察したところ、全てのクローンにてGFP陽性細胞が観察され、その陽性率は50~100%であった。 (2) Confirmation of transferred chromosomes (2-1) Observation with fluorescence microscope When nine cloned colonies were observed under a fluorescence microscope, GFP-positive cells were observed in all clones, and the positive rate was 50 to 100. %Met.
PCR解析で確認した4クローン(クローン名:hMSC-UC No.3 #1-01、#1-02、#2-01、#3-01)をHGF (hgf), GDNF (gdnf), IGF-1 (igf-1), 及びluciferase (e-luc)を保持するPACをプローブにしてFISH解析を行ったところ、核型異常を起こさず、ヒト人工染色体を保持するクローンが2クローンあることが確かめられた(図35、hMSC-UC No.3、#3-01)。 (2-2)
PCR解析で確認した上記4クローンについて、Okamotoら(BBRC, 295:354, 2002)の手法に従い、骨、軟骨、脂肪細胞へ分化誘導を行い、親株と同等の分化能を維持しているか否かを確認できる。 (3) In vitro differentiation induction The above 4 clones confirmed by PCR analysis were induced to differentiate into bone, cartilage, and adipocytes according to the method of Okamoto et al. (BBRC, 295: 354, 2002), and the differentiation potential equivalent to that of the parent strain was obtained. You can check whether it is maintained or not.
(1)CD4陽性CD45RB高陽性CD25陰性(CD4+CD45RBHigh+CD25-)T細胞の単離
8週齢BALB/cAJcl(日本クレア)の雌30匹を1週間の馴化後、麻酔下で心採血により脱血し、脾臓及び腸間膜リンパ節を採取した。脾臓はMACS system(Miltenyi Biotech)を用いて組織分散後に溶血処理した。腸間膜リンパ節は、1 mLシリンジ用プランジャーを用いて潰し、40μmセルストレーナーでろ過した。脾臓及び腸間膜リンパ節細胞を合わせ、CD4マイクロビーズ(Miltenyi Biotech)処理によりCD4陽性細胞を分離した後、CD4+CD45RBHigh+CD25-T細胞を単離するため、抗体(APC-H7 Rat anti-mouse CD4抗体(BD)、FITC Rat Anti-Mouse CD45RB抗体(BD)、PE/Cy7 anti-mouse CD25抗体(BioLegend))で標識し、フローサイトメーター(MoFlo XDP, BECKMAN)により分取した。 (Example 14) Verification of enteritis healing effect of immortalized MSC in an inflammatory bowel disease model (1) CD4 positive CD45RB high positive CD25 negative (CD4 + CD45RB High + CD25- ) Isolation of
9週齢雌のSCID(C.B-17/lcr-scid/scidJcl:日本クレア)に、1匹当たり4.0×105個のCD4+CD45RBHigh+CD25-T細胞を尾静脈注射により移植した(細胞移植群:1群8匹×5群、未処置群(PBS投与):1群×7匹)。移植後は、週3回の体重測定および毛並みと便状態の観察を実施した。細胞投与から21日後に体重変化に基づき、統計解析ソフトウェアJMPの「多変数によるブロック割り付け」システムを利用したランダム化割り付けを行い、細胞移植群を群分けした。その際、相対体重平均±2SDから外れた個体は除外した。 (2) CD4 + CD45RB High + CD25 --T cell transfer 9-week-old female SCID (CB-17 / lcr-scid / scidJcl: Japan Claire), 4.0 x 10 5 CD4 + CD45RB High + CD25 - T per animal Cells were transplanted by tail vein injection (cell transplant group: 1
移植用細胞を得るため、hMSC-UC(親MSC)及び不死化MSCを各細胞培養条件により培養した。 (3) Culture of transplanted cells In order to obtain cells for transplantation, hMSC-UC (parent MSC) and immortalized MSC were cultured under each cell culture condition.
CD4+CD45RB High+CD25-T細胞を移植後21日目、28日目、35日目に、親MSC、不死化MSCをマウス1匹当たり1.0×106個を尾静脈注射により投与した。ポジティブコントロールとして、Dexamethasone (Sigma): Dex(1 mg/kg、100μL/匹)を21日目から14日間、皮下投与した。Dex溶液は21日目及び28日目の群平均体重を基に用時調製した。 (4) MSC administration (therapeutic cell transplantation)
On the 21st, 28th, and 35th days after transplantation of CD4 + CD45RB High + CD25-T cells, parental MSC and immortalized MSC were administered 1.0 × 10 6 per mouse by tail vein injection. As a positive control, Dexamethasone (Sigma): Dex (1 mg / kg, 100 μL / animal) was subcutaneously administered from the 21st day to the 14th day. Dex solution was prepared at the time of use based on the group average body weight on the 21st and 28th days.
実験開始から42日目に、全採血し、血清を調製した(-80℃保存)。さらに幽門輪から肛門(結腸~直腸)間の消化管を採材し、状態の評価と重量及び長さの測定を実施後、10%ホルマリンで固定した。 (5) Collection On the 42nd day from the start of the experiment, all blood was collected and serum was prepared (stored at -80 ° C). Furthermore, the gastrointestinal tract between the pyloric ring and the anus (colon to rectum) was sampled, and after evaluation of the condition and measurement of weight and length, the gastrointestinal tract was fixed with 10% formalin.
本明細書で引用した全ての刊行物、特許及び特許出願をそのまま参考として本明細書に組み入れるものとする。 The present invention can be used in the fields of cell medicine and regenerative medicine. The cell immortalization technique of the present invention can be used not only for normal cells with slow cell proliferation but also for cancer cells and the like, and can also be used in the field of basic research. Furthermore, single-cell cloning, which is essential for gene transfer and chromosome transfer, is also possible. In addition, the cells obtained by the present invention continue to proliferate without aging, which facilitates quality control, promotes mechanization of culture, significantly reduces cell production costs, and handles a large number of cells. Is possible. Therefore, the present invention expands the range of applicable diseases in cell medicine and regenerative medicine, and contributes to the activation of domestic and foreign medical-related industries.
All publications, patents and patent applications cited herein are incorporated herein by reference in their entirety.
Claims (18)
- 以下の工程を含む、可逆的不死化細胞の製造方法。
(1)不死化遺伝子を搭載する染色体非組み込み型RNAウイルスベクターを哺乳類の細胞に導入し、該細胞で不死化遺伝子を発現させる工程、及び
(2)工程(1)で得られた細胞を培養し、増殖させる工程 A method for producing reversible immortalized cells, which comprises the following steps.
(1) A step of introducing a chromosome-non-integrating RNA viral vector carrying an immortalizing gene into mammalian cells and expressing the immortalizing gene in the cells, and (2) culturing the cells obtained in step (1). And the process of growing - 前記不死化遺伝子が、Bmi-1遺伝子、TERT遺伝子、及びSV40T遺伝子よりなる群から選択される1種又は2種以上の不死化遺伝子である、請求項1に記載の方法。 The method according to claim 1, wherein the immortalizing gene is one or more immortalizing genes selected from the group consisting of the Bmi-1 gene, the TERT gene, and the SV40T gene.
- 前記不死化遺伝子が以下の(a)~(d)のいずれかである、請求項1又は2に記載の方法。
(a) Bmi-1遺伝子、TERT遺伝子、及びSV40T遺伝子の組み合わせ
(b) Bmi-1遺伝子とTERT遺伝子の組み合わせ
(c) TERT遺伝子とSV40T遺伝子の組み合わせ
(d) TERT遺伝子 The method according to claim 1 or 2, wherein the immortalizing gene is any one of the following (a) to (d).
(a) Combination of Bmi-1 gene, TERT gene, and SV40T gene
(b) Combination of Bmi-1 gene and TERT gene
(c) Combination of TERT gene and SV40T gene
(d) TERT gene - 前記細胞が、体細胞である、請求項1~3のいずれか1項に記載の方法。 The method according to any one of claims 1 to 3, wherein the cell is a somatic cell.
- 前記体細胞が、体性幹細胞である、請求項4に記載の方法。 The method according to claim 4, wherein the somatic cell is a somatic stem cell.
- 前記体性幹細胞が、間葉系幹細胞である、請求項5に記載の方法。 The method according to claim 5, wherein the somatic stem cell is a mesenchymal stem cell.
- 前記染色体非組み込み型RNAウイルスベクターが、マイナス鎖RNAウイルスベクターである、請求項1~6のいずれか1項に記載の方法。 The method according to any one of claims 1 to 6, wherein the chromosomal non-integrated RNA viral vector is a negative-strand RNA viral vector.
- 前記マイナス鎖RNAウイルスベクターが、パラミクソウイルスベクターである、請求項7に記載の方法。 The method according to claim 7, wherein the negative strand RNA virus vector is a paramyxovirus vector.
- 前記パラミクソウイルスベクターが、センダイウイルスベクターである、請求項8に記載の方法。 The method according to claim 8, wherein the paramyxovirus vector is a Sendai virus vector.
- 前記センダイウイルスベクターが、温度感受性センダイウイルスベクターである、請求項9に記載の方法。 The method according to claim 9, wherein the Sendai virus vector is a temperature-sensitive Sendai virus vector.
- 前記染色体非組み込み型RNAウイルスベクターがセンダイウイルスベクターであり、前記工程(2)の培養後、該センダイウイルスベクターを除去する工程をさらに含む、請求項1に記載の方法。 The method according to claim 1, wherein the chromosomal non-integrating RNA virus vector is a Sendai virus vector, further comprising a step of removing the Sendai virus vector after culturing in the step (2).
- 前記センダイウイルスベクターの除去が、培養温度を35℃から37℃に変更することにより行われる、請求項11に記載の方法。 The method according to claim 11, wherein the removal of the Sendai virus vector is performed by changing the culture temperature from 35 ° C to 37 ° C.
- 前記工程(2)の培養後、不死化細胞をクローニングする工程をさらに含む、請求項1~12のいずれか1項に記載の方法。 The method according to any one of claims 1 to 12, further comprising a step of cloning immortalized cells after culturing in the step (2).
- 請求項1~13のいずれか1項に記載の方法により得られる不死化細胞。 Immortalized cells obtained by the method according to any one of claims 1 to 13.
- Bmi-1遺伝子、TERT遺伝子、及びSV40T遺伝子よりなる群から選択される1種又は2種以上の不死化遺伝子を搭載するセンダイウイルスベクターを除去可能な状態で含む、不死化細胞。 Immortalized cells containing a Sendai virus vector carrying one or more immortalizing genes selected from the group consisting of Bmi-1 gene, TERT gene, and SV40T gene in a removable state.
- 請求項14又は15に記載の不死化細胞を含む再生医療製品。 A regenerative medicine product containing the immortalized cell according to claim 14 or 15.
- Bmi-1遺伝子、TERT遺伝子、及びSV40T遺伝子よりなる群から選択される1種又は2種以上の不死化遺伝子を搭載する、温度感受性センダイウイルスベクター。 A temperature-sensitive Sendai virus vector carrying one or more immortalizing genes selected from the group consisting of Bmi-1 gene, TERT gene, and SV40T gene.
- 請求項17に記載の温度感受性センダイウイルスベクターを含む、可逆的不死化細胞作製用キット。 A kit for producing reversible immortalized cells, which comprises the temperature-sensitive Sendai virus vector according to claim 17.
Priority Applications (4)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US18/251,805 US20230407268A1 (en) | 2020-11-06 | 2021-11-05 | Method for producing reversibly immortalized cell |
EP21889268.5A EP4242317A1 (en) | 2020-11-06 | 2021-11-05 | Method for producing reversibly immortalized cell |
JP2022560822A JPWO2022097716A1 (en) | 2020-11-06 | 2021-11-05 | |
CN202180075178.0A CN116419968A (en) | 2020-11-06 | 2021-11-05 | Preparation method of reversible immortalized cells |
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
JP2020186146 | 2020-11-06 | ||
JP2020-186146 | 2020-11-06 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2022097716A1 true WO2022097716A1 (en) | 2022-05-12 |
Family
ID=81458329
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/JP2021/040757 WO2022097716A1 (en) | 2020-11-06 | 2021-11-05 | Method for producing reversibly immortalized cell |
Country Status (5)
Country | Link |
---|---|
US (1) | US20230407268A1 (en) |
EP (1) | EP4242317A1 (en) |
JP (1) | JPWO2022097716A1 (en) |
CN (1) | CN116419968A (en) |
WO (1) | WO2022097716A1 (en) |
Citations (9)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
JP2003174870A (en) * | 2001-10-12 | 2003-06-24 | Naoya Kobayashi | Immortalized bone marrow mesenchymal stem cell |
JP2009291157A (en) * | 2008-06-06 | 2009-12-17 | Hidetoshi Yamashita | Conjunctival epithelial cell line |
WO2010008054A1 (en) * | 2008-07-16 | 2010-01-21 | ディナベック株式会社 | Method for production of reprogrammed cell using chromosomally unintegrated virus vector |
WO2010084970A1 (en) * | 2009-01-23 | 2010-07-29 | 国立大学法人大阪大学 | Feeder cell for target cell induction |
WO2015046229A1 (en) | 2013-09-24 | 2015-04-02 | ディナベック株式会社 | Method for improving efficiency of inducing pluripotent stem cell |
WO2017078176A1 (en) | 2015-11-05 | 2017-05-11 | 株式会社Quarrymen&Co. | Immortalized stem cells, and method for producing same |
JP2017221219A (en) | 2005-10-18 | 2017-12-21 | ナショナル ジューイッシュ ヘルスNational Jewish Health) | Conditionally immortalized long-term stem cells and methods of making and using such cells |
JP2020186146A (en) | 2019-05-13 | 2020-11-19 | キヤノン株式会社 | Method for manufacturing glass and method for manufacturing optical element |
JP2021061773A (en) * | 2019-10-11 | 2021-04-22 | 国立研究開発法人農業・食品産業技術総合研究機構 | Methods for producing swine infectious viruses |
-
2021
- 2021-11-05 WO PCT/JP2021/040757 patent/WO2022097716A1/en active Application Filing
- 2021-11-05 EP EP21889268.5A patent/EP4242317A1/en active Pending
- 2021-11-05 JP JP2022560822A patent/JPWO2022097716A1/ja active Pending
- 2021-11-05 US US18/251,805 patent/US20230407268A1/en active Pending
- 2021-11-05 CN CN202180075178.0A patent/CN116419968A/en active Pending
Patent Citations (11)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
JP2003174870A (en) * | 2001-10-12 | 2003-06-24 | Naoya Kobayashi | Immortalized bone marrow mesenchymal stem cell |
JP3953399B2 (en) | 2001-10-12 | 2007-08-08 | 直哉 小林 | Immortalized bone marrow mesenchymal stem cells |
JP2017221219A (en) | 2005-10-18 | 2017-12-21 | ナショナル ジューイッシュ ヘルスNational Jewish Health) | Conditionally immortalized long-term stem cells and methods of making and using such cells |
JP2009291157A (en) * | 2008-06-06 | 2009-12-17 | Hidetoshi Yamashita | Conjunctival epithelial cell line |
WO2010008054A1 (en) * | 2008-07-16 | 2010-01-21 | ディナベック株式会社 | Method for production of reprogrammed cell using chromosomally unintegrated virus vector |
JP5763340B2 (en) | 2008-07-16 | 2015-08-12 | ディナベック株式会社 | Method for producing reprogrammed cells using a non-chromosomal viral vector |
WO2010084970A1 (en) * | 2009-01-23 | 2010-07-29 | 国立大学法人大阪大学 | Feeder cell for target cell induction |
WO2015046229A1 (en) | 2013-09-24 | 2015-04-02 | ディナベック株式会社 | Method for improving efficiency of inducing pluripotent stem cell |
WO2017078176A1 (en) | 2015-11-05 | 2017-05-11 | 株式会社Quarrymen&Co. | Immortalized stem cells, and method for producing same |
JP2020186146A (en) | 2019-05-13 | 2020-11-19 | キヤノン株式会社 | Method for manufacturing glass and method for manufacturing optical element |
JP2021061773A (en) * | 2019-10-11 | 2021-04-22 | 国立研究開発法人農業・食品産業技術総合研究機構 | Methods for producing swine infectious viruses |
Non-Patent Citations (5)
Title |
---|
B AN, H. ET AL.: "Efficient generation of transgene-free human induced pluripotent stem cells (iPSCs) by temperature-sensitive Sendai virus vectors", PNAS, vol. 108, no. 34, 23 August 2011 (2011-08-23), pages 14234 - 14239, XP002684868, Retrieved from the Internet <URL:https://www.pnas.org/content/108/34/14234> DOI: 10.1073/PNAS.1103509108 * |
BAN HNISHISHITA NFUSAKI NTABATA TSAEKI KSHIKAMURA MTAKADA NINOUE MHASEGAWA MKAWAMATA S, PROC NATL ACAD SCI U S A., vol. 108, no. 34, 23 August 2011 (2011-08-23), pages 14234 - 9 |
BAN HNISHISHITA NFUSAKI NTABATA TSAEKI KSHIKAMURA MTAKADA NINOUE MHASEGAWA MKAWAMATA S, PROC NATL ACAD SCI USA., vol. 108, no. 34, 23 August 2011 (2011-08-23), pages 14234 - 9 |
FANG-YING MENG ET AL.: "Reversible immortalization of human hepatocytes mediated by retroviral transfer and site-specific recombination", WORLD JOURNAL OF GASTROENTEROLOGY, vol. 20, no. 36, 1 September 2014 (2014-09-01), pages 13119 - 13126 |
OKAMOTO ET AL., BBRC, vol. 295, 2002, pages 354 |
Also Published As
Publication number | Publication date |
---|---|
CN116419968A (en) | 2023-07-11 |
EP4242317A1 (en) | 2023-09-13 |
US20230407268A1 (en) | 2023-12-21 |
JPWO2022097716A1 (en) | 2022-05-12 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
Sun et al. | Stem cell-based therapies for Duchenne muscular dystrophy | |
US10568911B2 (en) | Multipotent stem cells and uses thereof | |
Otsu et al. | Differentiation of induced pluripotent stem cells into dental mesenchymal cells | |
Narita et al. | Effects of transforming growth factor-beta 1 and ascorbic acid on differentiation of human bone-marrow-derived mesenchymal stem cells into smooth muscle cell lineage | |
EP2137300B1 (en) | Pluripotent autologous stem cells from oral or gastrointestinal mucosa | |
US8574567B2 (en) | Multipotent stem cells and uses thereof | |
JP2012509072A (en) | Reprogramming cells to a pluripotent state | |
JP2022513355A (en) | Induced pluripotent cells containing controllable transgenes for conditional immortality | |
US20070243608A1 (en) | Platelet bioreactor | |
WO2016072446A1 (en) | Virus vector, cell, and construct | |
WO2022097716A1 (en) | Method for producing reversibly immortalized cell | |
US20210284966A1 (en) | Method of differentiation into mesenchymal stem cells through continuous subculture of dedifferentiated stem cells | |
JP2010051326A (en) | Method of inducing differentiation of mesodermal stem cell into nervous system cell | |
JP2019076008A (en) | Precursor cells capable of differentiating into osteocytes, chondrocytes, adipocytes or neurons, method for producing the same, and method for using the same | |
CN115505573A (en) | Genetically engineered mesenchymal stem cell with enhanced migration activity and application thereof in stem cell treatment and stem cell cosmetology | |
JP2008119003A (en) | Method for inducing differentiation from mesodermal stem cell, embryonic stem cell or immortalized mesodermal stem cell to nervous system cell | |
JP2019103393A (en) | Method for induction of skeletal muscle stem cell | |
JP2019170393A (en) | Method for fabricating ips cell | |
WO2014038655A1 (en) | Method of producing intestinal epithelium-derived somatic stem cells | |
Liu et al. | Human telomerase reverse transcriptase gene-transfected effects on biological characteristics of Schwann cells | |
WO2014038653A1 (en) | Production method for kidney-derived somatic stem cells | |
Curini et al. | Nucleofection of Ovine Amniotic Fluid-Derived Mesenchymal Stem Cells |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 21889268 Country of ref document: EP Kind code of ref document: A1 |
|
ENP | Entry into the national phase |
Ref document number: 2022560822 Country of ref document: JP Kind code of ref document: A |
|
WWE | Wipo information: entry into national phase |
Ref document number: 18251805 Country of ref document: US |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
ENP | Entry into the national phase |
Ref document number: 2021889268 Country of ref document: EP Effective date: 20230606 |